US20220204580A1 - Polypeptides and uses thereof - Google Patents

Polypeptides and uses thereof Download PDF

Info

Publication number
US20220204580A1
US20220204580A1 US17/551,701 US202117551701A US2022204580A1 US 20220204580 A1 US20220204580 A1 US 20220204580A1 US 202117551701 A US202117551701 A US 202117551701A US 2022204580 A1 US2022204580 A1 US 2022204580A1
Authority
US
United States
Prior art keywords
aib
xaa
c18diacid
amide
cntatc
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/551,701
Inventor
Maria Aleksandra Bednarek
Sivaneswary GENAPATHY
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MedImmune Ltd
Original Assignee
MedImmune Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by MedImmune Ltd filed Critical MedImmune Ltd
Priority to US17/551,701 priority Critical patent/US20220204580A1/en
Publication of US20220204580A1 publication Critical patent/US20220204580A1/en
Assigned to MEDIMMUNE LIMITED reassignment MEDIMMUNE LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GENAPATHY, Sivaneswary, BEDNAREK, MARIA ALEKSANDRA
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the present invention relates to polypeptides which are pramlintide analogues and uses thereof.
  • the present invention relates to polypeptides which are pramlintide analogues conjugated to half-life extending moieties such as albumin binding moieties and uses thereof.
  • Pramlintide is a synthetic analogue of human amylin with three proline substitutions, at positions 25, 28 and 29. As a result of these substitutions, pramlintide has a reduced propensity to form amyloid fibrils, thereby overcoming a physicochemical liability of native human amylin (Kruger D F, Gloster M A. Pramlintide for the treatment of insulin-requiring diabetes mellitus: rationale and review of clinical data. Drugs. 2004; 64(13):1419-32).
  • Pramlintide is clinically used in amylin replacement therapies and simulates the important glucoregulatory actions of amylin. These glucoregulatory actions complement those of insulin by regulating the rate of appearance of glucose in the circulation, and are achieved through three primary mechanisms: slowing the rate of gastric emptying, suppression of post-meal glucagon secretion and suppression of food intake (Roth J D et. al. GLP-1R and amylin agonism in metabolic disease: complementary mechanisms and future opportunities. Br J Pharmacol. 2012; 166(1):121-136).
  • Pramlintide has been used as an adjunct to insulin in patients with diabetes who have failed to reach desired glucose control despite optimal insulin therapy (Pullman J, et. al. Pramlintide is used in the management of insulin-using patients with type 2 and type 1 diabetes. Vasc Health Risk Manag. 2006; 2(3):203-212).
  • the present invention relates to polypeptides that are pramlintide analogues conjugated to albumin binding moieties (e.g. lipids).
  • polypeptide or a pharmaceutically acceptable salt thereof, comprising the amino acid sequence:
  • Xaa 24 is Gly, Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
  • lipidated polypeptide or a pharmaceutically acceptable salt thereof, comprising the amino acid sequence:
  • a pharmaceutical composition comprising a polypeptide, a lipidated polypeptide or pharmaceutically acceptable salt of the invention and a pharmaceutically acceptable excipient.
  • a method for treating a disease or disorder in a subject comprising administering a polypeptide, a lipidated polypeptide, pharmaceutically acceptable salt or a pharmaceutical composition of the invention.
  • an article of manufacture comprising a polypeptide, a lipidated polypeptide, a pharmaceutically acceptable salt or a pharmaceutical composition of the invention.
  • kits comprising a polypeptide, a lipidated polypeptide, a pharmaceutically acceptable salt or a pharmaceutical composition of the invention, optionally further comprising instructions for use.
  • the present inventors have observed that pramlintide conjugated to an albumin binding moiety, such as a lipid, has poor stability (e.g. the fibril-forming tendency of pramlintide is increased) under conditions required for drug product formulation.
  • the present invention is based, at least in part, on the finding that the polypeptides (e.g. lipidated polypeptides) described herein may exhibit improved stability (e.g. reduced or no fibrillation tendency) as compared to such pramlintide conjugates.
  • the present inventors have found that when pramlintide is conjugated to a lipid to increase the half-life, the fibril-forming tendency also increases. Accordingly, the polypeptides (e.g. lipidated polypeptides) described herein may bring the benefit of extended half-life compared to pramlintide but without the fibril-forming tendency of alternative lipidated pramlintide analogues.
  • Peptides disclosed here can be formulated in or chemically conjugated to e.g. a protein, polymeric drug carrier or advance drug delivery system that enhances the chemical stability and or physical stability and or the circulatory exposure of the therapeutic moiety. The present inventors have further found that the polypeptides (e.g.
  • lipidated polypeptides described herein may exhibit improved physical and/or chemical stability as compared to human amylin or pramlintide. Furthermore, the polypeptides (e.g. lipidated polypeptides) described herein may have similar or improved selectivity to human amylin (hAMYR) compared to pramlintide.
  • hAMYR human amylin
  • amino acid positions of the polypeptides are numbered according to the corresponding position in pramlintide having the sequence set forth in SEQ ID NO. 1.
  • amino acids are referred to by their conventional three-letter or single-letter abbreviations (e.g. Ala or A for alanine, Arg or R for arginine, etc.). In the case of certain less common or non-naturally occurring amino acids (i.e.
  • amino acids other than the 20 encoded by the standard mammalian genetic code unless they are referred to by their full name, frequently employed three- or four-character codes are employed for residues thereof, including ⁇ MeSer ((S)-2-amino-2-methyl-3-phenylpropanoic acid), ⁇ MePhe ((S)-2-amino-2-methyl-3-phenylpropanoic acid), Aib (2-amino-2-methylpropanoic acid), Dab (2,4-diaminobutanoic acid) and ⁇ -Glu ( ⁇ -glutamic acid).
  • ⁇ MeSer ((S)-2-amino-2-methyl-3-phenylpropanoic acid)
  • ⁇ MePhe ((S)-2-amino-2-methyl-3-phenylpropanoic acid)
  • Aib (2-amino-2-methylpropanoic acid
  • Dab 2,4-diaminobutanoic acid
  • ⁇ -Glu
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides of the invention are isolated polypeptides (e.g. isolated lipidated polypeptides).
  • the polypeptides of the invention comprise at least one albumin binding moiety. Without being bound by theory, it is thought that the albumin binding moiety protects the polypeptide against clearance and degradation, thereby extending the half-life of the polypeptide.
  • albumin binding moiety refers to a compound that binds to albumin.
  • Exemplary albumin binding moieties suitable for use in the polypeptides of the invention include lipids (e.g. a fatty acid derivative), albumin-binding peptides, albumin-binding proteins, or small molecule ligands that bind to albumin.
  • the albumin binding moiety is a lipid, e.g. a lipid described herein.
  • polypeptides of the invention may comprise one or more albumin binding moiety (e.g. lipid), e.g. one, two or three albumin binding moieties. In preferred embodiments, the polypeptides of the invention comprise only one albumin binding moiety (e.g. lipid).
  • albumin binding moiety e.g. lipid
  • albumin binding moieties e.g. one, two or three albumin binding moieties.
  • the polypeptides of the invention comprise only one albumin binding moiety (e.g. lipid).
  • the albumin binding moiety may be attached to an amino acid residue of the polypeptide.
  • the albumin binding moiety e.g. lipid
  • the albumin binding moiety is attached to the amino acid residue through a linker.
  • the albumin binding moiety e.g. lipid
  • the albumin binding moiety may be attached to the amino acid residue via an ester, a sulfonyl ester, a thioester, an amide, an amine or a sulphonamide. Accordingly, it will be understood that the albumin binding moiety (e.g.
  • the linker includes an acyl group, a sulphonyl group, an N atom, an O atom or an S atom which forms part of the ester, sulphonyl ester, thioester, amide, amine or sulphonamide.
  • an acyl group in the albumin binding moiety (e.g. lipid) or the linker forms part of an amide or ester with the amino acid residue.
  • the albumin binding moiety e.g. lipid
  • the albumin binding moiety (e.g. lipid) may be attached to any residue at position Xaa ⁇ 4 to Xaa 37 (e.g. to the ⁇ N of a lysine residue) of the polypeptide.
  • the albumin binding moiety (e.g. lipid) is attached to the side chain of an amino acid residue in the polypeptide, for example to the ⁇ N of a lysine residue.
  • the albumin binding moiety (e.g. lipid) is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide).
  • the albumin binding moiety (e.g. lipid) is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide). In some embodiments, the albumin binding moiety (e.g. lipid) is attached to the amino acid residue at Xaa ⁇ 4, Xaa ⁇ 3, Xaa ⁇ 2, Xaa ⁇ 1 or Xaa 1 (e.g. to the ⁇ N of a lysine residue at Xaa ⁇ 4, Xaa ⁇ 3, Xaa ⁇ 2, Xaa ⁇ 1 or Xaa 1). In preferred embodiments, the albumin binding moiety (e.g. lipid) is attached to Xaa ⁇ 4, Xaa ⁇ 1 or Xaa 1 (either to the N-terminus or to the side chain of Xaa ⁇ 4, Xaa ⁇ 1 or Xaa 1).
  • the albumin binding moiety is a lipid.
  • the polypeptides of the invention may comprise at least one lipid (referred to herein as “lipidated polypeptide”). Without being bound by theory, it is thought that the lipid acts as an albumin binding moiety and protects the polypeptide against clearance and degradation, thereby extending the half-life of the polypeptide. The lipid may also modulate the potency of the compound as an agonist to the amylin (calcitonin) receptor.
  • the polypeptide comprises at least one lipidated amino acid residue. In some embodiments, the polypeptide comprises at least two lipidated amino acid residues. In preferred embodiments, the polypeptide contains only one lipidated amino acid residue.
  • the lipid may be attached to an amino acid residue of the polypeptide. In some embodiments, the lipid is attached to the amino acid residue through a linker (referred to herein as “linker-lipid”). In alternative embodiments, the lipid is directly attached to the amino acid residue without an intervening linker.
  • the lipid may be attached to the amino acid residue via an ester, a sulfonyl ester, a thioester, an amide, an amine or a sulphonamide.
  • the lipid or the linker includes an acyl group, a sulphonyl group, an N atom, an O atom or an S atom which forms part of the ester, sulphonyl ester, thioester, amide, amine or sulphonamide.
  • an acyl group in the lipid or linker forms part of an amide or ester with the amino acid residue.
  • the lipid is attached to an acylation site on the amino acid residue.
  • the lipid may be attached to any residue at position Xaa ⁇ 4 to Xaa 37 (e.g. to the ⁇ N of a lysine residue) of the polypeptide.
  • the lipid is attached to the side chain of an amino acid residue in the polypeptide, for example to the ⁇ N of a lysine residue.
  • the lipid is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide).
  • the lipid is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide). In some embodiments, the lipid is attached to the amino acid residue at Xaa ⁇ 4, Xaa ⁇ 3, Xaa ⁇ 2, Xaa ⁇ 1 or Xaa 1 (e.g. to the ⁇ N of a lysine residue at Xaa ⁇ 4, Xaa ⁇ 3, Xaa ⁇ 2, Xaa ⁇ 1 or Xaa 1).
  • the lipid is attached to Xaa ⁇ 4, Xaa ⁇ 1 or Xaa 1 (either to the N-terminus or to the side chain of Xaa ⁇ 4, Xaa ⁇ 1 or Xaa 1).
  • the lipid may comprise a hydrocarbon chain having from 10 to 26 C atoms, e.g. from 14 to 24 C atoms, e.g. from 16 to 22 C atoms.
  • the hydrocarbon chain may contain 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 C atoms.
  • the lipid has 18 to 20 C atoms.
  • the lipid may have 18 C atoms or 20 C atoms.
  • the hydrocarbon chain may be linear or branched, and may be saturated or unsaturated.
  • it can include a functional group at the end of the lipophilic chain, e.g. a carboxylic acid group which may or may not be protected during synthesis.
  • the lipid comprises a dicarboxylic acid.
  • the lipid may comprise C12diacid, C14diacid, C16diacid, C17diacid, C18diacid, C19diacid or C20diacid.
  • the lipid comprises C18diacid or C20diacid.
  • the albumin binding moiety (e.g. lipid) may be attached to the polypeptide through a linker.
  • the linker may comprise one or more residues of any naturally occurring or non-naturally occurring amino acid.
  • the linker may comprise a combination of residues, as single or repeating units.
  • the linker may comprise multiple combinations of residues, as single or repeating units, each of which may independently be a residue of Glu, y-Glu, Lys, ⁇ -Lys, Asp, ⁇ -Asp, Gaba, ⁇ -Ala (3-aminopropanoyl), O2Oc (2-(2-(2-aminoethoxy)ethoxy)acetic acid), PEG2 (3-(2-(2-aminoethoxy)ethoxy)propanoic acid), PEG4 (1-amino-3,6,9,12-tetraoxapentadecan-15-oic acid), PEG8 (1-amino-3,6,9,12,15,18,21,24-octaoxaheptacosan-27-oic acid, PEG12 (1-amino-3,6,9,12,15,18,21,24,27,30,33,36-dodecaoxanonatriacontan-39-oic acid
  • y-Glu and ⁇ -Asp refer to amino acids where the alpha-amino group and the side chain carboxyl group participate in peptide bond formation.
  • ⁇ -Lys refers to an amino acid where the epsilon-amino and carboxyl group of lysine participate in peptide bond formation.
  • the linker comprises a residue of ⁇ -Glu, e.g. ⁇ Glu, ⁇ Glu- ⁇ Glu, ⁇ Glu-(O2Oc)-(O2Oc) or ⁇ Glu-(PEG2)-(PEG2).
  • the linker consists of ⁇ Glu, ⁇ Glu- ⁇ Glu, ⁇ Glu-(O2Oc)-(O2Oc) or ⁇ Glu-(PEG2)-(PEG2).
  • the polypeptide comprises any one of the linker and lipid combinations set forth in any one of the rows in Table 2.
  • the linker may be attached to the amino acid residue via an ester, a sulfonyl ester, a thioester, an amide, an amine or a sulphonamide. Accordingly it will be understood that optionally the linker includes an acyl group, a sulphonyl group, an N atom, an O atom or an S atom which forms part of the ester, sulphonyl ester, thioester, amide, amine or sulphonamide. Optionally, an acyl group in the linker forms part of an amide or ester with the amino acid residue. Accordingly, in preferred embodiments the linker is attached to an acylation site on the amino acid residue.
  • the linker may be attached to a site (e.g. an acylation site) at the N-terminus of the lipidated polypeptide or to the ⁇ amino group “ ⁇ N” of a residue in the lipidated polypeptide, e.g. to ⁇ N of a lysine residue.
  • a site e.g. an acylation site
  • the polypeptide comprises a combination of linker, lipid and acylation site set forth in any one of the rows of Table 2.
  • the linker may be attached to any residue at position Xaa ⁇ 4 to Xaa 37 (e.g. to the ⁇ N of a lysine residue) of the polypeptide.
  • the linker is attached to the side chain of an amino acid residue in the polypeptide, for example to the ⁇ N of a lysine residue.
  • the linker is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide).
  • the linker is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide). In some embodiments, the linker is attached to the amino acid residue at Xaa ⁇ 4, Xaa ⁇ 3, Xaa ⁇ 2, Xaa ⁇ 1 or Xaa 1 (e.g. to the ⁇ N of a lysine residue at Xaa ⁇ 4, Xaa ⁇ 3, Xaa ⁇ 2, Xaa ⁇ 1 or Xaa 1).
  • the linker is attached to Xaa ⁇ 4, Xaa ⁇ 1 or Xaa 1 (either to the N-terminus or to the side chain of Xaa ⁇ 4, Xaa ⁇ 1 or Xaa 1).
  • the linker is attached to a site (e.g. an acylation site) selected from the N-terminus of the polypeptide, ⁇ N of a lysine at position Xaa (1) “1K”, the ⁇ N of a lysine at position Xaa ( ⁇ 1) “ ⁇ 1K”, or the ⁇ N of a lysine at position Xaa ( ⁇ 4) “ ⁇ 4K”.
  • a site e.g. an acylation site
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides (e.g. lipidated polypeptides) of the invention may comprise one or more amino acid modifications or substitutions compared to the pramlintide sequence [SEQ ID NO: 1].
  • the polypeptides comprises one or more non-proteinogenic amino acids.
  • Non-proteinogenic amino acids may include alpha methyl amino acids, D-enantiomers of naturally occurring amino acids, 2,4-diaminobutanoic acid (Dab), and (2S,4R)-4-hydroxypyrrolidine-2-carboxylic acid (Hyp).
  • the polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises one or more non-proteinogenic amino acids between positions 14-37, optionally at one or more of 14, 17 or 20-37.n some embodiments, the polypeptide (e.g.
  • lipidated polypeptide comprises one or more alpha methyl amino acids between positions 14-37, optionally at one or more of alpha methyl amino acids at positions 14, 17 or 20-37.
  • Polypeptides e.g. lipidated polypeptides
  • alpha methyl amino acids include 2-amino-2-methylpropanoic acid (Aib), alpha-methyl glutamine ( ⁇ MeGlu), alpha methyl phenylalanine ( ⁇ MePhe or ⁇ MeF), alpha-methyl leucine ( ⁇ MeLeu) and alpha-methyl serine ( ⁇ MeSer).
  • the alpha methyl amino acid can be Aib, ⁇ MeGlu, ⁇ MePhe, ⁇ MeLeu or ⁇ MeSer, or any combination thereof.
  • the polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises at least one alpha methyl amino acid, optionally selected from Aib, ⁇ MePhe and ⁇ MeSer.
  • the reference to ⁇ MePhe and ⁇ MeF herein refers to (S)-2-amino-2-methyl-3-phenylpropanoic acid.
  • the reference to ⁇ MeSer herein refers to (S)-2-amino-3-hydroxy-2-methylpropanoic acid.
  • the alpha methyl amino acid is Aib, ⁇ MePhe or ⁇ MeSer.
  • the polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises one or more non-proteinogenic amino acids between positions 14-37 selected from the group consisting of: 2,4-diaminobutanoic acid (Dab), (2S,4R)-4-hydroxypyrrolidine-2-carboxylic acid (Hyp), D-leucine (dL), D-isoleucine (dl) and D-proline (dP).
  • polypeptide e.g. lipidated polypeptide
  • the polypeptide does not comprise (2S)-2-aminohexanedioic acid) (Aad) and/or does not comprise Aad at positions 14-37.
  • the polypeptide does not comprise Aib at one or more of positions 15, 16, 17, 19 or 20.
  • the polypeptide comprises Aib at one or more of positions 15, 16, 17, 19 or 20 and at least one different non-proteinogenic amino acid (e.g. an alpha methyl amino acid that is not Aib) at positions 14-37.
  • the polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises one or more natural amino acid substitutions or modifications compared to the pramlintide sequence [SEQ ID NO: 1].
  • the polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises one or more of the following natural amino acid substitutions or modifications: deleted 1K (A1K), Ile 4, Ala 4, Glu 14, His 14, Trp 15, Arg 17, Ser 17, Glu 17, Pro 20, Ile 20, His 21, Ala 21, Glu 21, Gly 21, Lys 21, Pro 21, Arg 21, Ser 21, His 22, Pro 24, Ala 25, Arg 26, Ser 28, His 31, Glu 31, Pro 31, Arg 31, His 34, Pro 33, Pro 34, Glu 35, Arg 35, Pro 35 and Pro 37.
  • deleted 1K A1K
  • Ile 4 Ala 4
  • Glu 14, His 14, Trp 15 Arg 17, Ser 17, Glu 17, Pro 20
  • Ile 20 His 21, Ala 21, Glu 21, Gly 21, Lys 21, Pro 21, Arg 21, Ser 21, His 22, Pro 24, Ala 25, Arg 26, Ser 28, His 31, Glu 31, Pro 31, Arg 31, His 34, Pro 33, Pro 34, Glu 35, Arg 35, Pro 35 and Pro 37.
  • polypeptide e.g. lipidated polypeptides
  • polypeptide may comprise a combination of non-proteinogenic amino acids and natural amino acid substitutions or modifications compared to the pramlintide sequence [SEQ ID NO: 1].
  • polypeptide e.g. lipidated polypeptide
  • lipidated polypeptide that is a pramlintide analogue, or a pharmaceutically acceptable salt thereof, comprising any of the amino acid sequence modification combinations set forth in Table 3.
  • a polypeptide e.g. lipidated polypeptide
  • a pramlintide analogue or a pharmaceutically acceptable salt thereof, having an alpha methyl amino acid at position 23.
  • the alpha methyl amino acid is ⁇ MePhe.
  • polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises an alpha methyl amino acid (e.g. ⁇ MePhe) at position 23
  • the polypeptide e.g. lipidated polypeptide
  • a polypeptide e.g. lipidated polypeptide that is a pramlintide analogue, or a pharmaceutically acceptable salt thereof having at least two Aib residues.
  • the polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises Aib at at least two of positions 17 and 20-37.
  • the polypeptide e.g. lipidated polypeptide
  • polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises at least two Aib residues
  • the polypeptide comprises any one of the following combinations of modifications:
  • a polypeptide e.g. lipidated polypeptide
  • a pramlintide analogue or a pharmaceutically acceptable salt thereof, having an alpha methyl amino acid at position 21.
  • the alpha methyl amino acid is Aib.
  • polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises an alpha methyl amino acid (e.g. Aib) at position 21
  • the polypeptide comprises any one of the following combinations of modifications:
  • a polypeptide e.g. lipidated polypeptide
  • a pramlintide analogue or a pharmaceutically acceptable salt thereof, having an alpha methyl amino acid at position 17.
  • the alpha methyl amino acid is Aib.
  • polypeptide e.g. lipidated polypeptide
  • the polypeptide comprises an alpha methyl amino acid (e.g. Aib) at position 17
  • the polypeptide comprises any one of the following combinations of modifications:
  • polypeptides (e.g. lipidated polypeptides) of the invention may exhibit favourable pharmacokinetic properties as compared to pramlintide.
  • the polypeptides (e.g. lipidated polypeptides) of the invention may have an extended half-life as compared to pramlintide.
  • half-life is used to refer to the time taken for the concentration of isolated polypeptide in plasma to decline to 50% of its original level. Methods to determine the half-life of proteins are known in the art and are described in Example 4.
  • an extended half-life is advantageous, as it permits the therapeutic proteins to be administered according to a safe and convenient dosing schedule, e.g. lower doses that can be administered less frequently.
  • the achievement of lower doses may provide further advantages such as the provision of an improved safety profile.
  • pramlintide requires frequent and inconvenient administration.
  • the polypeptides (e.g. lipidated polypeptides) of the invention may have a half-life of at least 4 hours in rat models (see Example 4).
  • the polypeptide (e.g. lipidated polypeptide) has a half-life of at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 5 hours, at least 6 hours, at least 7 hours, at least 8 hours, at least 9 hours, at least 10 hours, at least 11 hours, at least 12 hours, at least 13 hours or at least 14 hours in rat models.
  • the polypeptide (e.g. lipidated polypeptide) has a half-life of at least 14 hours.
  • the polypeptides (e.g. lipidated polypeptides) of the invention may exhibit reduced tendency to undergo fibrillation in pharmaceutically relevant aqueous media, especially at pH values in the range from 4 to 7, as compared to lipidated pramlintide.
  • the polypeptide (e.g. lipidated polypeptide) exhibits reduced tendency to undergo fibrillation in pharmaceutically relevant aqueous media, especially at pH values in the range from 4 to 7, as compared to pramlintide which is lipidated in a similar manner e.g. the same lipid is attached, the lipid is attached through the same linker and/or the lipid is attached at the same position.
  • Exemplary lipidated pramlintide molecules are given in Table 1, for example SEQ ID NO. 3, 4, 5, 6, 7, 112 and 113.
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides of the invention may be suited for formulation in acidic media (e.g. pH 4) and in neutral or near-neutral media (e.g. pH 7 or 7.4).
  • acidic media e.g. pH 4
  • neutral or near-neutral media e.g. pH 7 or 7.4
  • Such polypeptides e.g. lipidated polypeptides
  • the polypeptide (e.g. lipidated polypeptide) shows no detectable fibrillation after about 5 hours, after about 7 hours, after about 9 hours, after about 11 hours, after about 13 hours, after about 15 hours, after about 17 hours or after about 20 hours at pH 4 and 37° C., e.g. under the conditions described in Example 3.
  • the polypeptide (e.g. lipidated polypeptide) shows no detectable fibrillation after about 48 hours, after about 72 hours, after about 96 hours, after about 108 hours, after about 120 hours, after 132 about hours or after about 144 hours at pH 4 and 37° C., e.g. under the conditions described in Example 3.
  • the polypeptide (e.g. lipidated polypeptide) shows no detectable fibrillation after 144 hours at pH 4 and 37° C., e.g. under the conditions described in Example 3.
  • the formation of fibrils is detected by an increase in fluorescence intensity in a Thioflavin T fibrillation assay, e.g. as described in Example 3.
  • the polypeptides (e.g. lipidated polypeptides) of the invention are soluble at concentrations required for therapeutic efficacy. In some embodiments, the lipidated polypeptides of the invention are soluble at a concentration of at least 1 mg/mL under the conditions described in Example 3.
  • polypeptides e.g. lipidated polypeptides
  • amylin receptor agonists i.e. they are capable of binding to, and inducing signalling by, one or more receptors or receptor complexes regarded as physiological receptors for human amylin.
  • receptors or receptor complexes regarded as physiological receptors for human amylin.
  • These include the human calcitonin receptor hCTR, as well as complexes comprising the human calcitonin receptor hCTR and at least one of the human receptor activity modifying proteins designated hRAMP1, hRAMP2 and hRAMP3.
  • Complexes between hCTR and hRAMP1, hRAMP2 and hRAMP3 are designated hAMYR1, hAMYR2 and hAMYR3 (i.e.
  • a compound is considered an amylin receptor agonist if it has agonist activity at one or more of hAMYR1, hAMYR2 and hAMYR3.
  • a compound may be considered an amylin receptor agonist if it has agonist activity at hAMYR3.
  • the ability to induce cAMP formation as a result of binding to the relevant receptor or receptor complex is typically regarded as indicative of agonist activity.
  • Other intracellular signaling pathways or events may also be used as readouts for amylin receptor agonist activity. These may include calcium release, arrestin recruitment, receptor internalization, kinase activation or inactivation, lipase activation, inositol phosphate release, diacylglycerol release or nuclear transcription factor translocation.
  • EC50 values may be used as a measure of agonist potency at a given receptor.
  • An EC50 value is a measure of the concentration of a compound required to achieve half of that compound's maximal activity in a particular assay, for example a cAMP assay as described in Example 2.
  • the present inventors have shown that certain polypeptides (e.g. lipidated polypeptides) disclosed herein exhibit greater or similar selectivity to hAMYR over hCTR as pramlintide, as measured using cAMP release from binding to hAMYR and hCTR.
  • Pramlintide exhibits at least 10-fold selectivity to hAMYR as compared to hCTR.
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides (e.g. lipidated polypeptides) of the invention may exhibit improved efficacy, e.g. as amylin receptor agonists, as compared to lipidated pramlintide.
  • the polypeptide (e.g. lipidated polypeptide) has at least about 1-fold selectivity to hAMYR over hCTR, optionally at least about 2-fold, at least about 4-fold, at least about 6-fold, at least about 8-fold, at least about 10-fold, at least about 12-fold, at least about 14-fold, at least about 16-fold, at least about 18-fold, at least about 20-fold, at least about 50-fold, at least about 75-fold, or at least about 100-fold selectivity to hAMYR over hCTR.
  • the polypeptide (e.g. lipidated polypeptide) has at least about 10-fold selectivity to hAMYR over hCTR.
  • the polypeptide e.g. lipidated polypeptide
  • the polypeptide has around 12-20 fold, around 14-18 fold, optionally around 16-fold selectivity to hAMYR over hCTR.
  • the isolated polypeptide has an EC50 measured under the conditions described in Example 2 (i.e. containing 0.1% bovine serum albumin (BSA)) of below about 1.4 nM, below about 1.2 nM, below about 1 nM, below about 0.8 nM, below about 0.6 nM, below about 0.4nM, below about 0.3 nM, or below about 0.2 nM.
  • BSA bovine serum albumin
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides (e.g. lipidated polypeptides) of the invention may be chemically stable, e.g. they may form in a formulation an acceptable percentage of degradation products produced over a defined period of time by chemical pathways, such as deamidation, aggregation, or oxidation.
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides (e.g. lipidated polypeptides) of the invention may be chemically conjugated to a protein or polymeric drug carrier, or formulated in an advance drug delivery system, that enhances the chemical stability and/or physical stability and/or the circulatory exposure of the polypeptide.
  • polypeptide or a pharmaceutically acceptable salt thereof wherein the polypeptide comprises any one of the lipid linkers as set forth in Table 2 and any one of the sequence modifications as set forth in Table 3.
  • polypeptide or a pharmaceutically acceptable salt thereof wherein the polypeptide comprises the lipid linker and amino acid sequence modification combinations set forth in Table 4.
  • polypeptides (e.g. lipidated polypeptides) of the invention may be produced by any method known in the art. The production of polypeptides such as amylin or analogues thereof is well known in the art.
  • the polypeptide (e.g. lipidated polypeptides) of the invention can thus be produced by chemical synthesis, e.g. solid phase polypeptide synthesis using t-Boc or Fmoc chemistry, or other well-established techniques. They may alternatively be produced by recombinant expression of a nucleic acid molecule encoding a fusion polypeptide in a host cell. Following synthesis, the polypeptides (e.g. lipidated polypeptides) of the invention may optionally be isolated or purified.
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides of the invention are provided in a pharmaceutical composition.
  • compositions of the invention may comprise one or more excipient(s).
  • excipients are known in the art, see for instance Remington's Pharmaceutical Sciences (by Joseph P. Remington, 18th ed., Mack Publishing Co., Easton, Pa.), which is incorporated herein in its entirety.
  • the present invention encompasses therapies which involve administering the polypeptides (e.g. lipidated polypeptides) of the invention to an animal, in particular a mammal, for instance a human, for preventing, treating, or ameliorating symptoms associated with a disease, disorder, or infection.
  • a mammal for instance a human
  • the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention may be used in therapy, for example for treating a disease or disorder.
  • a method of treating a disease or disorder comprising administering to a subject or patient in need thereof a therapeutically effective amount of the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention.
  • the use or method may comprise administering a therapeutically effective schedule that has less frequent doses of the polypeptides (e.g. lipidated polypeptides) of the invention than the therapeutically effective dosing schedule of pramlintide.
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides of the invention may be used in the treatment and/or prevention of obesity, metabolic diseases such as diabetes (e.g. type 1 or type 2 diabetes), and/or obesity-related conditions.
  • the polypeptides (e.g. lipidated polypeptides) of the invention may be used in a method of treating obesity, overweight, morbid obesity, obesity prior to surgery, obesity-linked inflammation, obesity-linked gallbladder disease, sleep apnea and respiratory problems, hyperlipidemia, degeneration of cartilage, osteoarthritis, or reproductive health complications of obesity or overweight such as infertility in a subject, the method comprising administering a therapeutically effective amount of the polypeptide (e.g. lipidated polypeptide) to the subject.
  • a therapeutically effective amount of the polypeptide e.g. lipidated polypeptide
  • the polypeptide e.g. lipidated polypeptide
  • Metabolic diseases that may be treated by the polypeptide (e.g. lipidated polypeptide) of the invention include diabetes, type 1 diabetes, type 2 diabetes, gestational diabetes, pre-diabetes, insulin resistance, impaired glucose tolerance (IGI), disease states associated with elevated blood glucose levels, metabolic disease including metabolic syndrome, or hyperglycemia e.g. abnormal postprandial hyperglycemia.
  • diabetes type 1 diabetes, type 2 diabetes, gestational diabetes, pre-diabetes, insulin resistance, impaired glucose tolerance (IGI), disease states associated with elevated blood glucose levels, metabolic disease including metabolic syndrome, or hyperglycemia e.g. abnormal postprandial hyperglycemia.
  • IGI impaired glucose tolerance
  • polypeptides e.g. lipidated polypeptides
  • the polypeptides of the invention are used for the treatment of type 1 diabetes or type 2 diabetes.
  • polypeptides e.g. lipidated polypeptides
  • pharmaceutical compositions of the invention may be used for treating, inhibiting or reducing weight gain, promoting weight loss, reducing food intake, and/or reducing excess body weight.
  • polypeptides e.g. lipidated polypeptides
  • pharmaceutical compositions of the invention may be used in the treatment and/or prevention of an eating disorder, Alzheimer's disease, hepatic steatosis (“fatty liver”), kidney failure, arteriosclerosis (e.g. atherosclerosis), cardiovascular disease, macrovascular disease, microvascular disease, diabetic heart (including diabetic cardiomyopathy and heart failure as a diabetic complication), coronary heart disease, peripheral artery disease or stroke, cancer, dumping syndrome, hypertension e.g. pulmonary hypertension, or dyslipidemia e.g. atherogenic dyslipidemia, cholescystitis, or short bowel syndrome.
  • an eating disorder e.g. hepatic steatosis (“fatty liver”), kidney failure, arteriosclerosis (e.g. atherosclerosis), cardiovascular disease, macrovascular disease, microvascular disease, diabetic heart (including diabetic cardiomyopathy and heart failure as a diabetic complication), coronary heart disease, peripheral artery disease or
  • polypeptides e.g. lipidated polypeptides
  • the route of administration of polypeptides (e.g. lipidated polypeptides) of the invention, or pharmaceutical compositions thereof can be, for example, oral, parenteral, by inhalation or topical.
  • the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition thereof is administered by parenteral administration to a subject or patient.
  • parenteral as used herein includes, e.g., intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, rectal, or vaginal administration.
  • the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition thereof is administered by injection, such as by intravenous, subcutaneous or intramuscular injection, to a subject or patient.
  • the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition thereof is administered by subcutaneous injection.
  • Administration by injection such as by subcutaneous injection, offers the advantage of better comfort for the subject or patient and the opportunity to administer to a subject or patient outside of a hospital setting.
  • the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition thereof is administered by self-administration.
  • the subject or patient is a mammal, in particular a human.
  • the polypeptide or pharmaceutical composition is administered to the subject in combination with insulin.
  • the present invention provides an article of manufacture comprising the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention.
  • the present invention provides a kit comprising the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention.
  • the kit may comprise a package containing the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition, optionally with instructions.
  • the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention are formulated in single dose vials or a container closure system (e.g. pre-filled syringe).
  • a container closure system e.g. pre-filled syringe
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • nucleic acid sequences are written left to right in 5′ to 3′ orientation; amino acid sequences are written left to right in amino to carboxy orientation, respectively.
  • “About” may generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, and more typically, within 5% of a given value or range of values. Optionally, the term “about” shall be understood herein as plus or minus ( ⁇ ) 5%, optionally ⁇ 4%, ⁇ 3%, ⁇ 2%, ⁇ 1%, ⁇ 0.5%, ⁇ 0.1%, of the numerical value of the number with which it is being used.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government, or listed in the U.S. Pharmacopeia, European Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • polypeptides e.g. lipidated polypeptides
  • methods described herein will be apparent to a person of skill in the art.
  • pramlintide analogue peptides were synthesized as C-terminal carboxamides using rink amide MBHA resin (100-200 mesh). All peptides were prepared by automated synthesis using a Liberty BlueTM microwave solid phase peptide synthesizer (CEM Corporation, N.C., USA) using the Fmoc/tBu protocol. Manufacturer-supplied protocols were applied for coupling of amino acids in DMF and deprotection of Fmoc protecting group using piperidine in DMF (20% v/v). Asparagine, cysteine, glutamine and histidine were incorporated as their sidechain trityl (Trt) derivatives.
  • Trt sidechain trityl
  • Lysine was incorporated as the sidechain tert-butyloxycarbonyl (Boc) derivative.
  • Serine, threonine and tyrosine were incorporated as sidechain tert-butyl (tBu) ethers, and aspartate and glutamate as their sidechain OtBu esters.
  • Arginine was incorporated as the sidechain 2,2,4,6,7-pentamethyldihydrobenzofuran-5-sulfonyl (Pbf) derivative.
  • Boc-Lys(Fmoc) was incorporated when a subsequent chemical modification of the N-terminal lysine side chain was required.
  • coupling of an albumin binding moiety, such as a lipid was performed manually using HATU as a coupling reagent in the presence of DI PEA.
  • Peptides were cleaved from the solid support by treatment with a mixture of TFA:TIS:EDT:thioanisole:water (90:2.5:2.5:2.5:2.5 v/v) for 4 h with agitation at room temperature. Thereafter, the cleavage mixtures were filtered, concentrated in vacuo, precipitated and washed with diethyl ether and solids were isolated by centrifugation. The linear crude peptides were dried under a flow of nitrogen and dissolved in 20% MeCN in water (v/v) with 1% TFA (v/v) and filtered.
  • the crude linear peptides were purified using a preparative RP-HPLC on a Varian SD-1 Prep Star binary pump system, monitoring by UV absorption at 210 nm using an Xbridge C18-A stationary phase (19.0 ⁇ 250 mm, 5 micron) column eluting a linear solvent gradient of 25-70% MeCN (0.1% TFA v/v) in water (0.1% TFA v/v) over 25 min.
  • the linear purified peptides were cyclised by treatment with iodine (1% w/v in methanol) for 10 min at room temperature and excess iodine was reduced by treatment with ascorbic acid (1% w/v in water).
  • the cyclic crude peptides were re-purified as described above.
  • the purified fractions were pooled, frozen and lyophilised.
  • LC/MS characterisation of purified peptides were performed on a Waters MassLynx 3100 platform using a XBridge C18 stationary phase (4.6 ⁇ 100 mm, 3 micron) eluting a linear binary gradient of 10-90% MeCN (0.1% TFA v/v) in water (0.1% TFA v/v) over 10 minutes at 1.5 mL/min at ambient temperature.
  • Analytes were detected by both UV absorption at 210 nm and ionization using a Waters 3100 mass detector (ESI+ mode).
  • Analytical RP-HPLC characterisation was performed on an Agilent 1260 Infinity system using an Agilent Polaris C8-A stationary phase (4.6 ⁇ 100 mm, 3 micron) eluting a linear binary gradient of 10-90% MeCN (0.1% TFA v/v) in water (0.1% TFA v/v) at 1.5 mL/min over 15 minutes at 40° C.
  • lipidated pramlintide analogue peptides such as cAMP production
  • hCTR human calcitonin receptor
  • RAMP3 human amylin receptor
  • rat CTR rat calcitonin receptor
  • RAM P3 receptor activity modifying protein
  • Cryopreserved cell stock was thawed rapidly in a water-bath, suspended in assay buffer (0.1% BSA (Sigma #A3059) in HBSS (Sigma #H8264) with 25 mM HEPES, pH 7.4 and containing 0.5 mM IB MX (Sigma#17018)) and spun at 240 ⁇ g for 5 minutes.
  • Cells were re-suspended in assay buffer at a batch-dependent optimized concentration (e.g. hCTR cells at 0.125 ⁇ 10 5 cells/mL, hAMYR3 cells at 0.125 ⁇ 10 5 cells/mL, rat CTR cells at at 1 ⁇ 10 5 cells/mL, rat AMYR3 at 2 ⁇ 10 5 cells/mL).
  • test peptide stock was prepared in DMSO and diluted in assay buffer to reach stated concentrations and transferred in duplicates into a 384-black shallow well microtitre assay plate (Corning #3676). Cells were added to the assay plate, incubated at room temperature for 30 minutes and the cAMP level measured using cAMP dynamic 2 HTRF kit (Cisbio, Cat #62AM4PEJ), following the two step protocol as per manufacturer's recommendations. The plates were read on an Envision (Perkin Elmer) using excitation wavelength of 320 nm and emission wavelengths of 620 nm & 665 nm.
  • EC50 values were transformed to % Delta F as described in the manufacturer's guidelines and analyzed as percent activation of maximal amylin or calcitonin effect by 4-parameter logistic fit to determine EC 50 values.
  • the selectivity of a peptide to hAMYR vs hCTR is defined as a ratio of EC50 values at the two receptors.
  • Thioflavin T (ThT) fibrillation assay is a useful tool to assess the aggregation kinetics of a peptide or protein under accelerated and stressed conditions that can be used to forecast the long-term viability of a compound in solution.
  • ThT can selectively bind amyloid fibrils and the resultant complex emits strong fluorescence signal at 482 nm when excited at 450 nm (Anal Biochem. 1989 March; 177(2):244-9). Monitoring of the change in fluorescence signal is an established method to study the fibril forming potential of peptides and proteins.
  • ThT purchased from Sigma Aldrich
  • ThT stock solution is prepared by dissolving the ThT powder in Milli-Q water and filtered to obtain a 0.25 mM solution.
  • the concentration of the solution is measured at 412 nm using an extinction coefficient of 36 mM ⁇ 1 cm ⁇ 1 .
  • Test peptides were dissolved at 1 mg/mL in 25 mM sodium acetate buffer pH 4.0.
  • the fibril forming potential of the test peptides was determined by measuring the average time taken to detect an increase in baseline corrected fluorescence intensity. A time>144 h indicates no increase in fluorescence intensity, relative to baseline, during the course of the experiment.
  • Conjugating pramlintide to a lipid increases the fibril-forming tendency as seen in Table 7.
  • Tht fibrillation assay of lipidated pramlintide analogues Time taken to detect increase Peptide in fluorescence intensity (h) 3 ⁇ 5 4 ⁇ 5 5 7 6 15 8 7 9 7 10 >144 11 >144 12 >144 13 25 15 >144 17 >144 18 >144 19 >144 20 >144 21 >144 24 >144 25 >144 28 45 33 >144 40 >144 44 >144 48 >144 66 >144 70 >144 80 >144 103 ⁇ 5 104 ⁇ 5 112 ⁇ 5 113 ⁇ 5 114 ⁇ 5 115 ⁇ 5 129 >144 156 >144
  • PK studies were to determine the plasma pharmacokinetic profile of lipidated pramlintide analogue peptides in fasted male SD rats after single intravenous (IV) and subcutaneous (SC) administration. PK studies were performed to determine the half-life (T 1/2 ) of test peptides. T 1/2 describes the time taken for the maximum plasma concentration (Cmax) of a test substance to halve its steady-state concentration when in circulation.
  • mice Male SD rats were purchased from Si Bei Fu Laboratory Animal Technology Co. Ltd (China). The animals were 6-8 weeks old with body weights of 200-300 g on the dosing date. The animals were housed in a 12-hour light/12-hour dark cycle environment and were fasted overnight before dosing. The body weight of the animals were recorded before dosing, 24 h and 48 h post dosing. Animals had free access to food and drinks, and the food consumption was quantified every day.
  • Test articles were administered at 20 nmol/kg. Blood samples were collected from each animal via Jugular vein. The sampling timepoints are as below.
  • the blood samples were transferred into eppendorf low binding tube containing K 2 EDTA. Above 0.150 mL blood were collected at each time point. Blood samples were centrifuged at 4,000 g for 5 minutes at 4° C. to obtain plasma. The plasma samples were stored frozen at ⁇ 75 ⁇ 15° C. until analysis.
  • Rats Male Sprague Dawley rats were obtained from Taconic Denmark, ApS at approximately 7 weeks of age. Rats were implanted with a microchip for identification, housed 4/cage with enrichment, free access to food and water, and allowed one week acclimatisation while non-invasive characterization was performed. Rats were on a 12:12 light:dark cycle that switches at 1 pm:1 am. Food intake was monitored via the HM2 system (Lafayette Instrument) that allows for monitoring in a home cage. As each rat enters an access tunnel to feed, an IR beam is broken, and the implanted microchip is read. Resulting changes to food weight is then assigned to the specific animal. Social order has shown no impact to overall feeding patterns and amounts.
  • the lipidated polypeptides show marked suppression of food intake compared to pramlintide.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Diabetes (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)

Abstract

Disclosed are polypeptides which are pramlintide analogues and uses thereof. In particular, the present invention relates to polypeptides of SEQ ID NO 2 which are pramlintide analogues conjugated to half-life extending moieties such as albumin binding moieties and uses thereof.

Description

    SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Dec. 15, 2021, is named 201085-WO-PCT_SL.txt and is 134,852 bytes in size.
  • FIELD OF THE INVENTION
  • The present invention relates to polypeptides which are pramlintide analogues and uses thereof. In particular, the present invention relates to polypeptides which are pramlintide analogues conjugated to half-life extending moieties such as albumin binding moieties and uses thereof.
  • BACKGROUND
  • Pramlintide is a synthetic analogue of human amylin with three proline substitutions, at positions 25, 28 and 29. As a result of these substitutions, pramlintide has a reduced propensity to form amyloid fibrils, thereby overcoming a physicochemical liability of native human amylin (Kruger D F, Gloster M A. Pramlintide for the treatment of insulin-requiring diabetes mellitus: rationale and review of clinical data. Drugs. 2004; 64(13):1419-32).
  • Pramlintide is clinically used in amylin replacement therapies and simulates the important glucoregulatory actions of amylin. These glucoregulatory actions complement those of insulin by regulating the rate of appearance of glucose in the circulation, and are achieved through three primary mechanisms: slowing the rate of gastric emptying, suppression of post-meal glucagon secretion and suppression of food intake (Roth J D et. al. GLP-1R and amylin agonism in metabolic disease: complementary mechanisms and future opportunities. Br J Pharmacol. 2012; 166(1):121-136). Pramlintide has been used as an adjunct to insulin in patients with diabetes who have failed to reach desired glucose control despite optimal insulin therapy (Pullman J, et. al. Pramlintide is used in the management of insulin-using patients with type 2 and type 1 diabetes. Vasc Health Risk Manag. 2006; 2(3):203-212).
  • Pharmacokinetic studies show that the terminal half-life of amylin in rats is around 13 minutes, and the half-life for pramlintide in human is ˜20-45 minutes (Roth J D et. al. GLP-1R and amylin agonism in metabolic disease: complementary mechanisms and future opportunities. Br J Pharmacol. 2012; 166(1):121-136).
  • There remains a need for pramlintide analogues which retain amylin agonist activity and provide advantages such as extended half-life and reduced fibrillation tendency.
  • SUMMARY OF INVENTION
  • The present invention relates to polypeptides that are pramlintide analogues conjugated to albumin binding moieties (e.g. lipids).
  • Thus, in one aspect, there is provided a polypeptide, or a pharmaceutically acceptable salt thereof, comprising the amino acid sequence:
      • Xaa (−4)-Xaa (−3)-Xaa (−2)-Xaa (−1)-Xaa 1-Cys 2-Asn 3-Xaa 4-Ala 5-Thr 6-Cys 7-Ala 8-Thr 9-Gln 10-Arg 11-Leu 12-Ala 13-Xaa 14-Xaa 15-Xaa 16-Xaa 17-His 18-Ser 19-Xaa 20-Xaa 21-Xaa 22-Xaa 23-Xaa 24-Xaa 25-Xaa 26-Xaa 27-Xaa 28-Xaa 29-Thr 30-Xaa 31-Xaa 32-Xaa 33-Xaa 34-Xaa 35-Xaa 36-Xaa 37-amide [SEQ ID NO:2], wherein:
      • Xaa (−4) is Lys(albumin binding moiety) or is absent;
      • Xaa (−3) is Gly or is absent;
      • Xaa (−2) is Gly or is absent;
      • Xaa (−1) is Gly, (albumin binding moiety), Lys(albumin binding moiety) or is absent;
      • Xaa 1 is Lys, Lys(albumin binding moiety), (albumin binding moiety) or is absent;
      • Xaa 4 is Thr, Ile or Ala;
      • Xaa 14 is Asn, His, Glu, 2,4-diaminobutanoic acid (Dab), or an alpha methyl amino acid;
      • Xaa 15 is Phe or Trp;
      • Xaa 16 is Leu or D-Leu (dL);
      • Xaa 17 is Val, Ser, Glu, Arg, (2S,4R)-4-hydroxypyrrolidine-2-carboxylic acid (Hyp), Dab or an alpha methyl amino acid (e.g. 2-amino-2-methylpropanoic acid [Aib]);
      • Xaa 20 is Ser, Ile, Pro or an alpha methyl amino acid (e.g. (S)-2-amino-3-hydroxy-2-methylpropanoic acid [αMeSer]);
      • Xaa 21 is Asn, Dab, His, Pro, Ser, Arg, Lys, Gly, Glu, Ala, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 22 is Asn, His, Hyp, Dab or an alpha methyl amino acid (e.g. Aib);
      • Xaa 23 is Phe, Hyp or an alpha methyl amino acid (e.g. (S)-2-amino-2-methyl-3-phenylpropanoic acid [αMePhe]);
  • Xaa 24 is Gly, Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 25 is Pro, Ala, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 26 is Ile, D-Ile (dl), Arg, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 27 is Leu, dL, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 28 is Pro, D-Pro (dP), Ser, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 29 is Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 31 is Asn, Glu, His, Arg, Pro, Dab or an alpha methyl amino acid (e.g. Aib);
      • Xaa 32 is Val, Hyp, Dab or an alpha methyl amino acid (e.g. Aib);
      • Xaa 33 is Gly, Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 34 is Ser, Pro, His, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 35 is Asn, Pro, Arg, Glu, Dab, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 36 is Thr, Hyp or an alpha methyl amino acid (e.g. Aib); and
      • Xaa 37 is Tyr, Pro, Hyp or an alpha methyl amino acid (e.g. Aib),
        and wherein the polypeptide comprises at least one albumin binding moiety.
  • In another aspect, there is provided a lipidated polypeptide, or a pharmaceutically acceptable salt thereof, comprising the amino acid sequence:
      • Xaa (−4)-Xaa (−3)-Xaa (−2)-Xaa (−1)-Xaa 1-Cys 2-Asn 3-Xaa 4-Ala 5-Thr 6-Cys 7-Ala 8-Thr 9-Gln 10-Arg 11-Leu 12-Ala 13-Xaa 14-Xaa 15-Xaa 16-Xaa 17-His 18-Ser 19-Xaa 20-Xaa 21-Xaa 22-Xaa 23-Xaa 24-Xaa 25-Xaa 26-Xaa 27-Xaa 28-Xaa 29-Thr 30-Xaa 31-Xaa 32-Xaa 33-Xaa 34-Xaa 35-Xaa 36-Xaa 37-amide [SEQ ID NO: 159], wherein:
      • Xaa (−4) is Lys(linker-lipid) or is absent;
      • Xaa (−3) is Gly or is absent;
      • Xaa (−2) is Gly or is absent;
      • Xaa (−1) is Gly, (linker-lipid), Lys(linker-lipid) or is absent;
      • Xaa 1 is Lys, Lys(linker-lipid), (linker-lipid) or is absent;
      • Xaa 4 is Thr, Ile or Ala;
      • Xaa 14 is Asn, His, Glu, 2,4-diaminobutanoic acid (Dab), or an alpha methyl amino acid;
      • Xaa 15 is Phe or Tip;
      • Xaa 16 is Leu or D-Leu (dL);
      • Xaa 17 is Val, Ser, Glu, Arg, (2S,4R)-4-hydroxypyrrolidine-2-carboxylic acid (Hyp), Dab or an alpha methyl amino acid (e.g. 2-amino-2-methylpropanoic acid [Aib]); Xaa 20 is Ser, Ile, Pro or an alpha methyl amino acid (e.g. (S)-2-amino-3-hydroxy-2-methylpropanoic acid [αMeSer]);
      • Xaa 21 is Asn, Dab, His, Pro, Ser, Arg, Lys, Gly, Glu, Ala, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 22 is Asn, His, Hyp, Dab or an alpha methyl amino acid (e.g. Aib);
      • Xaa 23 is Phe, Hyp or an alpha methyl amino acid (e.g. (S)-2-amino-2-methyl-3-phenylpropanoic acid [αMePhe]);
      • Xaa 24 is Gly, Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 25 is Pro, Ala, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 26 is Ile, D-Ile (dl), Arg, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 27 is Leu, dL, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 28 is Pro, D-Pro (dP), Ser, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 29 is Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 31 is Asn, Glu, His, Arg, Pro, Dab or an alpha methyl amino acid (e.g. Aib);
      • Xaa 32 is Val, Hyp, Dab or an alpha methyl amino acid (e.g. Aib);
      • Xaa 33 is Gly, Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 34 is Ser, Pro, His, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 35 is Asn, Pro, Arg, Glu, Dab, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 36 is Thr, Hyp or an alpha methyl amino acid (e.g. Aib);
      • Xaa 37 is Tyr, Pro, Hyp or an alpha methyl amino acid (e.g. Aib).
  • In yet another aspect, there is provided a polypeptide as set forth in Table 4.
  • In yet another aspect, there is provided a pharmaceutical composition comprising a polypeptide, a lipidated polypeptide or pharmaceutically acceptable salt of the invention and a pharmaceutically acceptable excipient.
  • In another aspect, there is provided a method for treating a disease or disorder in a subject comprising administering a polypeptide, a lipidated polypeptide, pharmaceutically acceptable salt or a pharmaceutical composition of the invention.
  • In a further aspect, there is provided a method for the production of a polypeptide or a lipidated polypeptide described herein.
  • In a further aspect, there is provided an article of manufacture comprising a polypeptide, a lipidated polypeptide, a pharmaceutically acceptable salt or a pharmaceutical composition of the invention.
  • In a further aspect, there is provided a kit comprising a polypeptide, a lipidated polypeptide, a pharmaceutically acceptable salt or a pharmaceutical composition of the invention, optionally further comprising instructions for use.
  • Aspects and embodiments of the invention are set out in the appended claims. These and other aspects and embodiments of the invention are also described herein.
  • BRIEF DESCRIPTION OF SEQUENCE LISTING
  • TABLE 1
    Compound Sequence Listing
    SEQ
    ID NO. Full sequence
      1 K[CNTATC]ATQRLANFLVHSSNNFGPIL
    (Pramlintide) PPTNVGSNTY-amide
      3 C18diacid-γE-K[CNTATC]ATQRLAN
    FLVHSSNNFGPILPPTNVGSNTY-amide
      4 C18diacid-γE-γE-GGG-K[CNTATC]
    ATQRLANFLVHSSNNFGPILPPTNVGSNT
    Y-amide
      5 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSSNNFGPILPPTNVGSNTY-
    amide
      6 K(O2Oc-O2Oc-γE-C18diacid)
    [CNTATC]ATQRLANFLVHSSNNFGPILP
    PTNVGSNTY-amide
      7 K(O2Oc-O2Oc-γE-C18diacid)GGGK
    [CNTATC]ATQRLANFLVHSSNNFGPILP
    PTNVGSNTY-amide
      8 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Dab)NFPAILSPTNVGSN
    TY-amide
      9 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFPAILSPTNVGSN
    TY-amide
     10 C18diacid-γE-[CNTATC]ATQRLAEF
    LRHSSNN(αMePhe)GPILPPTNVGSNT
    Y-amide
     11 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLAEFLRHSSNN(αMePhe)GPILPPTNV
    GSNTY-amide
     12 K(γE-C18diacid)K[CNTATC]ATQRL
    AEFLRHSSNN(αMePhe)GPILPPTNVGS
    NTY-amide
     13 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSSNN(αMePhe)GPILPPTNV
    GSNTY-amide
     14 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSSNN(αMePhe)GPILPPTNVGS
    NTY-amide
     15 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLRHSSNN(αMePhe)GPILPPTEVGS
    NTY-amide
     16 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLRHSSNN(αMePhe)GPILPPTEVGSN
    TY-amide
     17 K(O2Oc-O2Oc-γE-C18diacid)
    [CNTATC]ATQRLANFLRHSSNN
    (αMePhe)GPILPPTEVGSNTY-amide
     18 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLRHSSNN(αMePhe)GPILPPTEV
    GSNTY-amide
     19 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLRHSSNN(αMePhe)GPILPPTNVGSN
    TY-amide
     20 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLRHSSNN(αMePhe)GPILPPTNV
    GSNTY-amide
     21 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSSNN(αMePhe)GPILPPTNV
    GSRTY-amide
     22 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHS(αMeSer)NNFGPILPPTNV
    GSNTY-amide
     23 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSSNN(αMePhe)GPILPPTEV
    GSNTY-amide
     24 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSSNN(αMePhe)GPILPPTRV
    GSNTY-amide
     25 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSSNN(αMePhe)GPILPPTRVGSN
    TY-amide
     26 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFL(Aib)HSSNN(αMePhe)GPILP
    PTNVGSNTY-amide
     27 K(γE-γE-C18diacid)[CNIATC]ATQ
    RLANFLVHSS(Dab)NFGPILPPTNVGSR
    TY-amide
     28 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Dab)NFGPILPPTEVGSNT
    Y-amide
     29 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Dab)NFGPILPPTNVGSNT
    Y-amide
     30 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSS(Dab)NFGPILPPTEVG
    SNTY-amide
     31 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Dab)NFG(Aib)ILPPTNVG
    SNTY-amide
     32 K(γE-C18diacid)K[CNTATC]ATQRL
    A(Dab)FLVHSSNN(αMePhe)GPILPPT
    EVGSNTY-amide
     33 K(γE-C18diacid)K[CNTATC]ATQRL
    A(Dab)FLVHSSNNFGPILPPTNVGSNT
    Y-amide
     34 K(γE-C18diacid)K[CNTATC]ATQRL
    A(Dab)FLVHSSNNFGPILPPTEVGSNT
    Y-amide
     35 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNVGSNT
    Y-amide
     36 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSS(Aib)NFGPILPPTNVG
    SNTY-amide
     37 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLSHSS(Aib)NFGPILPPTNVGSNT
    Y-amide
     38 K(γE-CI8diacid)K[CNTATC]ATQRL
    AEFLVHSS(Aib)NFGPILPPTNVGSNT
    Y-amide
     39 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLEHSS(Aib)NFGPILPPTNVGSNT
    Y-amide
     40 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTHVGSNT
    Y-amide
     41 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTEVGSNT
    Y-amide
     42 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNVGSET
    Y-amide
     43 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLRHSS(Aib)NFGPILPPTEVGSNT
    Y-amide
     44 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLRHSS(Aib)NFGPILPPTEVGSNTY-
    amide
     45 K(O2Oc-O2Oc-γE-C18diacid)
    [CNTATC]ATQRLANFLRHSS(Aib)NFG
    PILPPTEVGSNTY-amide
     46 K(γE-C18diacid)K[CNTATC]ATQRL
    AHFLVHSS(Aib)NFGPILPPTNVGSNT
    Y-amide
     47 K(γE-C18diacid)K[CNTATC]ATQRL
    AHFLVHSS(Aib)NFGPILPPTNVGSET
    Y-amide
     48 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLRHSS(Aib)NFGPILPPTNVGSNTY-
    amide
     49 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFGPILPPTEVGSN
    TY-amide
     50 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLRHSS(Aib)NFGPILPPTEVGSN
    TY-amide
     51 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLSHSS(Aib)NFGPILPPTHVGSNT
    Y-amide
     52 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLSHSS(Aib)NFGPILPPTRVGSNT
    Y-amide
     53 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLSHSS(Aib)NFGPILPPTPVGSNT
    Y-amide
     54 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLSHSS(Aib)NFGPILPPTNVPSNT
    Y-amide
     55 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLSHSS(Aib)NFGPILPPTNVGSPT
    Y-amide
     56 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSS(Aib)NFGPILPPTNVGSNTY-
    amide
     57 K(γE-γE-C18diacid)K[CNTATC]AT
    QRLANFLVHSS(Aib)NFGPILPPTNVGS
    NTY-amide
     58 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNVGSNT
    P-amide
     59 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSS(Aib)NFGPI(dL)PPTNVGSN
    TY-amide
     60 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSS(Aib)NFGPIL(dP)PTNVGSN
    TY-amide
     61 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSS(Aib)NFGP(dI)LPPTNVGSN
    TY-amide
     62 K(γE-C18diacid)[CNTATC]ATQRLA
    NF(dL)VHSS(Aib)NFGPILPPTNVGSN
    TY-amide
     63 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSS(Aib)NFGPILPPTRVGSNTY-
    amide
     64 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSS(Aib)NFGPILPPTNVGSRTY-
    amide
     65 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLRHSS(Aib)NFGPILPPTNVGSNT
    Y-amide
     66 K(γE-γE-C18diacid)K[CNTATC]AT
    QRLANFLRHSS(Aib)NFGPILPPTNVGS
    NTY-amide
     67 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFGPILPPTRVGSN
    TY-amide
     68 (C18diacid-γE-[CNTATC]ATQRLAN
    FLVHSS(Aib)NFGPILPPTRVGSNTY-
    amide
     69 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTRVGSNT
    Y-amide
     70 K(γE-γE-C18diacid)K[CNTATC]AT
    QRLANFLVHSS(Aib)NFGPILPPTRVGS
    NTY-amide
     71 C18diacid-γE-K[CNTATC]ATQRLAN
    FLVHSS(Aib)NFGPILPPTRVGSNTY-
    amide
     72 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFGPILPPTNVGSR
    TY-amide
     73 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNVGSRT
    Y-amide
     74 K(γE-γE-C18diacid)K[CNTATC]AT
    QRLANFLVHSS(Aib)NFGPILPPTNVGS
    RTY-amide
     75 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLRHSS(Aib)NFGPILPPTNVGSN
    TY-amide
     76 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLRHSS(Aib)NFGPILPPTRVGSNTY-
    amide
     77 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLRHSS(Aib)NFGPILPPTRVGSN
    TY-amide
     78 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGP(Aib)LPPTNVG
    SNTY-amide
     79 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPI(Aib)PPTNVG
    SNTY-amide
     80 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPT(Aib)VG
    SNTY-amide
     81 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNV(Aib)
    SNTY-amide
     82 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNVGS
    (Aib)TY-amide
     83 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNVGSN
    (Aib)Y-amide
     84 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNVG
    (Aib)NTY-amide
     85 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPILPPTNVGSNT
    (Aib)-amide
     86 K(γE-C18diacid)K[CNTATC]ATQRL
    AHFL(Aib)HSS(Aib)NFGPILPPTEVG
    SNTY-amide
     87 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSS(Aib)NFGPILPPTNVG
    SNTP-amide
     88 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSS(Aib)NFGPIL(Aib)PTNVG
    SNTY-amide
     89 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFGPILPPT(Aib)
    VGSNTY-amide
     90 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLRHSS(Aib)NFGPILPPT(Aib)
    VGSNTY-amide
     91 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFGPILPPTNVGS
    (Aib)TY-amide
     92 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLRHSS(Aib)NFGPILPPTNVGS
    (Aib)TY-amide
     93 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSS(Dab)NFG(Aib)ILPP
    TNVGSNTY-amide
     94 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGP(Aib)LPPTNVG
    SNTY-amide
     95 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPI(Aib)PPTNVG
    SNTY-amide
     96 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPIL(Aib)PTNVG
    SNTY-amide
     97 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILP(Aib)TNVG
    SNTY-amide
     98 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPT(Aib)VG
    SNTY-amide
     99 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTN(Aib)G
    SNTY-amide
    100 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNV(Aib)
    SNTY-amide
    101 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNVG
    (Aib)NTY-amide
    102 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNVGS
    (Aib)TY-amide
    103 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSSNNFGPI(dL)PPTNVGSNTY-
    amide
    104 K(γE-C18diacid)[CNTATC]ATQRLA
    NFLVHSSNNFGPIL(dP)PTNVGSNTY-
    amide
    105 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLSHSS(Dab)NFG(Aib)ILPPTNVG
    SNTY-amide
    106 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSSNNFGP(Aib)LPPTNVGSNT
    Y-amide
    107 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLRHSSNNFGP(Aib)LPPTNVGSNT
    Y-amide
    108 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSSNNFGPI(Aib)PPTNVGSNT
    Y-amide
    109 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSSN(Aib)FGPILPPTNVGSNT
    Y-amide
    110 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSSNNF(Aib)PILPPTNVGSNT
    Y-amide
    111 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFLVHSSNHFGPILPPTNVGSETY-
    amide
    112 C20diacid-γE-K[CNTATC]ATQRLAN
    FLVHSSNNFGPILPPTNVGSNTY-amide
    113 C20diacid-γE-O2Oc-O2Oc-K
    [CNTATC]ATQRLANFLVHSSNNFGPILP
    PTNVGSNTY-amide
    114 K(γE-γE-C20diacid)[CNTATC]ATQ
    RLANFLVHSSNNFGPILPPTNVGSRTY-
    amide
    115 K(γE-γE-C20diacid)[CNTATC]ATQ
    RLANFLVHSSPNFPAILSPTNVGSNTY-
    amide
    116 K(γE-γE-C20diacid)[CNTATC]ATQ
    RLAEFLRHSSNNFGPILPPTNVGSNTY-
    amide
    117 K(γE-γE-C20diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFPAILSPTNVGSN
    TY-amide
    118 K(γE-γE-C20diacid)[CNIATC]ATQ
    RLANFLVHSIANFGPILPPTNVGSRTY-
    amide
    119 K(γE-γE-C20diacid)[CNTATC]ATQ
    RLANFLVHSPPNFPAILSPTNVGSNTY-
    amide
    120 K(γE-γE-C20diacid)[CNAATC]ATQ
    RLANWLVHSSPNFPAILSPTNVGSNTY-
    amide
    121 K(γE-γE-C20diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NF(Hyp)AILSPTN
    VGSNTY-amide
    122 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFPAILSPT(Dab)
    VGSNTY-amide
    123 K(γE-γE-C20diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFPAILSPT(Dab)
    VGSNTY-amide
    124 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFPAILSPTNVGS
    (Dab)TY-amide
    125 K(γE-γE-C20diacid)[CNTATC]ATQ
    RLANFLVHSS(Aib)NFPAILSPTNVGS
    (Dab)TY-amide
    126 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSS(Dab)NFGPILPPTNVG
    SNTY-amide
    127 K(γE-γE-C18diacid)[CNTATC]ATQ
    RLANFL(Aib)HSSNNFGPILPPTNVGSN
    TY-amide
    128 K(γE-C18diacid)K[CNTATC]ATQRL
    AEFL(Aib)HSSNNFGPILPPTNVGSNT
    Y-amide
    129 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNVGSNT
    Y-amide
    130 K(γE-C18diacid)K[CNTATC]ATQRL
    AEFL(Aib)HSSHNFGPILPPTNVGSNT
    Y-amide
    131 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSHNFGPILPPTNVGSNT
    Y-amide
    132 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSPNFGPILPPTNVGSNT
    Y-amide
    133 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSSNFGPILPPTNVGSNT
    Y-amide
    134 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTPVGSNT
    Y-amide
    135 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNHFGPILPPTNVGSNT
    Y-amide
    136 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNVGSNT
    P-amide
    137 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSRNFGPILPPTNVGSNT
    Y-amide
    138 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSPNFGPILPPTEVGSNT
    Y-amide
    139 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSPNFGPILPPTNVGSET
    Y-amide
    140 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTEVGSNT
    Y-amide
    141 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNVGSRT
    Y-amide
    142 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNVGSET
    Y-amide
    143 K(γE-C18diacid)GGK[CNTATC]ATQ
    RLANFL(Aib)HSSNNFGPILPPTNVGSN
    TY-amide
    144 K(γE-C18diacid)[CNTATC]ATQRLA
    HFL(Aib)HSSNNFGPILPPTNVGSNTY-
    amide
    145 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNVGHNT
    Y-amide
    146 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTHVGSET
    Y-amide
    147 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNHFGPILPPTNVGSET
    Y-amide
    148 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPILPPTNVGPNT
    Y-amide
    149 K(γE-C18diacid)[CNTATC]ATQRLA
    NFL(Aib)HSSNNFGPILPPTNVGSNTY-
    amide
    150 K(γE-C18diacid)[CNTATC]ATQRLA
    NFL(Aib)HSS(Dab)NFGPILPPTNVGS
    NTY-amide
    151 K(γE-C18diacid)[CNTATC]ATQRLA
    NFL(Aib)HSSNNFGPI(dL)PPTNVGSN
    TY-amide
    152 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSNNFGPRLPPTNVGSNT
    Y-amide
    153 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSKNFGPILPPTNVGSNT
    Y-amide
    154 K(γE-C18diacid)K[CNTATC]ATQRL
    ANFL(Aib)HSSGNFGPILPPTNVGSNT
    Y-amide
    155 K(γE-C18diacid)[CNTATC]ATQRLA
    NFL(Aib)HSSNNFGPILPPTRVGSNTY-
    amide
    156 K(γE-C18diacid)K[CNTATC]ATQRL
    A(Dab)FL(Aib)HSSNNFGPILPPTEVG
    SNTY-amide
    157 (C18diacid)K[CNTATC]ATQRLANFL
    VHSS(Aib)NFGPILPPT(Aib)VGSNT
    Y-amide
    158 K(C18diacid)[CNTATC]ATQRLANFL
    VHSS(Aib)NFGPILPPT(Aib)VGSNT
    Y-amide
    Table 1: The square bracket [] between the two cysteine residues (cys 2 and cys 7) indicate the presence of an intramolecular disulphide bridge.
  • DETAILED DESCRIPTION
  • The present inventors have observed that pramlintide conjugated to an albumin binding moiety, such as a lipid, has poor stability (e.g. the fibril-forming tendency of pramlintide is increased) under conditions required for drug product formulation. The present invention is based, at least in part, on the finding that the polypeptides (e.g. lipidated polypeptides) described herein may exhibit improved stability (e.g. reduced or no fibrillation tendency) as compared to such pramlintide conjugates.
  • For example, the present inventors have found that when pramlintide is conjugated to a lipid to increase the half-life, the fibril-forming tendency also increases. Accordingly, the polypeptides (e.g. lipidated polypeptides) described herein may bring the benefit of extended half-life compared to pramlintide but without the fibril-forming tendency of alternative lipidated pramlintide analogues. Peptides disclosed here can be formulated in or chemically conjugated to e.g. a protein, polymeric drug carrier or advance drug delivery system that enhances the chemical stability and or physical stability and or the circulatory exposure of the therapeutic moiety. The present inventors have further found that the polypeptides (e.g. lipidated polypeptides) described herein may exhibit improved physical and/or chemical stability as compared to human amylin or pramlintide. Furthermore, the polypeptides (e.g. lipidated polypeptides) described herein may have similar or improved selectivity to human amylin (hAMYR) compared to pramlintide.
  • Throughout this specification, amino acid positions of the polypeptides (e.g. lipidated polypeptides) are numbered according to the corresponding position in pramlintide having the sequence set forth in SEQ ID NO. 1.
  • Throughout this specification, amino acids are referred to by their conventional three-letter or single-letter abbreviations (e.g. Ala or A for alanine, Arg or R for arginine, etc.). In the case of certain less common or non-naturally occurring amino acids (i.e. amino acids other than the 20 encoded by the standard mammalian genetic code), unless they are referred to by their full name, frequently employed three- or four-character codes are employed for residues thereof, including αMeSer ((S)-2-amino-2-methyl-3-phenylpropanoic acid), αMePhe ((S)-2-amino-2-methyl-3-phenylpropanoic acid), Aib (2-amino-2-methylpropanoic acid), Dab (2,4-diaminobutanoic acid) and γ-Glu (γ-glutamic acid).
  • In embodiments of any aspect of the invention, the polypeptides (e.g. lipidated polypeptides) of the invention are isolated polypeptides (e.g. isolated lipidated polypeptides).
  • Albumin Binding Moiety
  • The polypeptides of the invention comprise at least one albumin binding moiety. Without being bound by theory, it is thought that the albumin binding moiety protects the polypeptide against clearance and degradation, thereby extending the half-life of the polypeptide. As used herein, “albumin binding moiety” refers to a compound that binds to albumin. Exemplary albumin binding moieties suitable for use in the polypeptides of the invention include lipids (e.g. a fatty acid derivative), albumin-binding peptides, albumin-binding proteins, or small molecule ligands that bind to albumin. Optionally, the albumin binding moiety is a lipid, e.g. a lipid described herein.
  • The polypeptides of the invention may comprise one or more albumin binding moiety (e.g. lipid), e.g. one, two or three albumin binding moieties. In preferred embodiments, the polypeptides of the invention comprise only one albumin binding moiety (e.g. lipid).
  • The albumin binding moiety (e.g. lipid) may be attached to an amino acid residue of the polypeptide. In some embodiments, the albumin binding moiety (e.g. lipid) is attached to the amino acid residue through a linker. In alternative embodiments, the albumin binding moiety (e.g. lipid) is directly attached to the amino acid residue without an intervening linker. The albumin binding moiety (e.g. lipid) may be attached to the amino acid residue via an ester, a sulfonyl ester, a thioester, an amide, an amine or a sulphonamide. Accordingly, it will be understood that the albumin binding moiety (e.g. lipid) or the linker includes an acyl group, a sulphonyl group, an N atom, an O atom or an S atom which forms part of the ester, sulphonyl ester, thioester, amide, amine or sulphonamide. Optionally, an acyl group in the albumin binding moiety (e.g. lipid) or the linker forms part of an amide or ester with the amino acid residue. Accordingly, in preferred embodiments the albumin binding moiety (e.g. lipid) is attached to an acylation site on the amino acid residue.
  • The albumin binding moiety (e.g. lipid) may be attached to any residue at position Xaa −4 to Xaa 37 (e.g. to the ϵN of a lysine residue) of the polypeptide. In some embodiments, the albumin binding moiety (e.g. lipid) is attached to the side chain of an amino acid residue in the polypeptide, for example to the ϵN of a lysine residue. In some embodiments, the albumin binding moiety (e.g. lipid) is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide).
  • In some embodiments, the albumin binding moiety (e.g. lipid) is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide). In some embodiments, the albumin binding moiety (e.g. lipid) is attached to the amino acid residue at Xaa −4, Xaa −3, Xaa −2, Xaa −1 or Xaa 1 (e.g. to the ϵN of a lysine residue at Xaa −4, Xaa −3, Xaa −2, Xaa −1 or Xaa 1). In preferred embodiments, the albumin binding moiety (e.g. lipid) is attached to Xaa −4, Xaa −1 or Xaa 1 (either to the N-terminus or to the side chain of Xaa −4, Xaa −1 or Xaa 1).
  • Lipid
  • In preferred embodiments, the albumin binding moiety is a lipid. Accordingly, the polypeptides of the invention may comprise at least one lipid (referred to herein as “lipidated polypeptide”). Without being bound by theory, it is thought that the lipid acts as an albumin binding moiety and protects the polypeptide against clearance and degradation, thereby extending the half-life of the polypeptide. The lipid may also modulate the potency of the compound as an agonist to the amylin (calcitonin) receptor.
  • In some embodiments, the polypeptide comprises at least one lipidated amino acid residue. In some embodiments, the polypeptide comprises at least two lipidated amino acid residues. In preferred embodiments, the polypeptide contains only one lipidated amino acid residue. The lipid may be attached to an amino acid residue of the polypeptide. In some embodiments, the lipid is attached to the amino acid residue through a linker (referred to herein as “linker-lipid”). In alternative embodiments, the lipid is directly attached to the amino acid residue without an intervening linker. The lipid may be attached to the amino acid residue via an ester, a sulfonyl ester, a thioester, an amide, an amine or a sulphonamide. Accordingly, it will be understood that the lipid or the linker includes an acyl group, a sulphonyl group, an N atom, an O atom or an S atom which forms part of the ester, sulphonyl ester, thioester, amide, amine or sulphonamide. Optionally, an acyl group in the lipid or linker forms part of an amide or ester with the amino acid residue. Accordingly, in preferred embodiments the lipid is attached to an acylation site on the amino acid residue.
  • The lipid may be attached to any residue at position Xaa −4 to Xaa 37 (e.g. to the ϵN of a lysine residue) of the polypeptide. In some embodiments, the lipid is attached to the side chain of an amino acid residue in the polypeptide, for example to the ϵN of a lysine residue. In some embodiments, the lipid is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide).
  • In some embodiments, the lipid is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide). In some embodiments, the lipid is attached to the amino acid residue at Xaa −4, Xaa −3, Xaa −2, Xaa −1 or Xaa 1 (e.g. to the ϵN of a lysine residue at Xaa −4, Xaa −3, Xaa −2, Xaa −1 or Xaa 1). In preferred embodiments, the lipid is attached to Xaa −4, Xaa −1 or Xaa 1 (either to the N-terminus or to the side chain of Xaa −4, Xaa −1 or Xaa 1).
  • In embodiments of any aspect of the invention, the lipid may comprise a hydrocarbon chain having from 10 to 26 C atoms, e.g. from 14 to 24 C atoms, e.g. from 16 to 22 C atoms. For example, the hydrocarbon chain may contain 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 C atoms. In preferred embodiments, the lipid has 18 to 20 C atoms. In particular, the lipid may have 18 C atoms or 20 C atoms. The hydrocarbon chain may be linear or branched, and may be saturated or unsaturated. Furthermore, it can include a functional group at the end of the lipophilic chain, e.g. a carboxylic acid group which may or may not be protected during synthesis.
  • Optionally, the lipid comprises a dicarboxylic acid. For example, the lipid may comprise C12diacid, C14diacid, C16diacid, C17diacid, C18diacid, C19diacid or C20diacid. In preferred embodiments, the lipid comprises C18diacid or C20diacid.
  • Linker
  • The albumin binding moiety (e.g. lipid) may be attached to the polypeptide through a linker. In embodiments of any aspect of the invention, the linker may comprise one or more residues of any naturally occurring or non-naturally occurring amino acid. The linker may comprise a combination of residues, as single or repeating units. For example, the linker may comprise multiple combinations of residues, as single or repeating units, each of which may independently be a residue of Glu, y-Glu, Lys, ϵ-Lys, Asp, β-Asp, Gaba, β-Ala (3-aminopropanoyl), O2Oc (2-(2-(2-aminoethoxy)ethoxy)acetic acid), PEG2 (3-(2-(2-aminoethoxy)ethoxy)propanoic acid), PEG4 (1-amino-3,6,9,12-tetraoxapentadecan-15-oic acid), PEG8 (1-amino-3,6,9,12,15,18,21,24-octaoxaheptacosan-27-oic acid, PEG12 (1-amino-3,6,9,12,15,18,21,24,27,30,33,36-dodecaoxanonatriacontan-39-oic acid). y-Glu and β-Asp refer to amino acids where the alpha-amino group and the side chain carboxyl group participate in peptide bond formation. ϵ-Lys refers to an amino acid where the epsilon-amino and carboxyl group of lysine participate in peptide bond formation.
  • In some embodiments, the linker comprises a residue of γ-Glu, e.g. γGlu, γGlu-γGlu, γGlu-(O2Oc)-(O2Oc) or γGlu-(PEG2)-(PEG2). In some embodiments, the linker consists of γGlu, γGlu-γGlu, γGlu-(O2Oc)-(O2Oc) or γGlu-(PEG2)-(PEG2).
  • In some embodiments of any aspect of the invention, the polypeptide comprises any one of the linker and lipid combinations set forth in any one of the rows in Table 2.
  • The linker may be attached to the amino acid residue via an ester, a sulfonyl ester, a thioester, an amide, an amine or a sulphonamide. Accordingly it will be understood that optionally the linker includes an acyl group, a sulphonyl group, an N atom, an O atom or an S atom which forms part of the ester, sulphonyl ester, thioester, amide, amine or sulphonamide. Optionally, an acyl group in the linker forms part of an amide or ester with the amino acid residue. Accordingly, in preferred embodiments the linker is attached to an acylation site on the amino acid residue.
  • The linker may be attached to a site (e.g. an acylation site) at the N-terminus of the lipidated polypeptide or to the ϵ amino group “ϵN” of a residue in the lipidated polypeptide, e.g. to ϵN of a lysine residue.
  • In some embodiments, the polypeptide comprises a combination of linker, lipid and acylation site set forth in any one of the rows of Table 2.
  • TABLE 2
    Combinations of linker, lipid and polypeptide acylation site
    Acylation
    Lipid Linker site Formula
    C18diacid γE-γE N- terminal
    Figure US20220204580A1-20220630-C00001
    C18diacid γE (εN)K
    Figure US20220204580A1-20220630-C00002
    C18diacid γE- (O2Oc)- (O2Oc) (εN)K
    Figure US20220204580A1-20220630-C00003
    C18diacid γE-γE (εN)K
    Figure US20220204580A1-20220630-C00004
    C20diacid γE-γE N- terminal
    Figure US20220204580A1-20220630-C00005
    C20diacid γE N- terminal
    Figure US20220204580A1-20220630-C00006
    C20diacid γE- (O2Oc)- (O2Oc) N- terminal
    Figure US20220204580A1-20220630-C00007
    C20diacid γE-γE (εN)K
    Figure US20220204580A1-20220630-C00008
    C18diacid Nil (εN)K
    Figure US20220204580A1-20220630-C00009
    C18diacid Nil N- terminal
    Figure US20220204580A1-20220630-C00010
  • The linker may be attached to any residue at position Xaa −4 to Xaa 37 (e.g. to the ϵN of a lysine residue) of the polypeptide. In some embodiments, the linker is attached to the side chain of an amino acid residue in the polypeptide, for example to the ϵN of a lysine residue. In some embodiments, the linker is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide).
  • In some embodiments, the linker is attached to the N-terminus of the polypeptide, (e.g. to a lysine at the N-terminus of the polypeptide). In some embodiments, the linker is attached to the amino acid residue at Xaa −4, Xaa −3, Xaa −2, Xaa −1 or Xaa 1 (e.g. to the ϵN of a lysine residue at Xaa −4, Xaa −3, Xaa −2, Xaa −1 or Xaa 1). In preferred embodiments, the linker is attached to Xaa −4, Xaa −1 or Xaa 1 (either to the N-terminus or to the side chain of Xaa −4, Xaa −1 or Xaa 1).
  • In some embodiments, the linker is attached to a site (e.g. an acylation site) selected from the N-terminus of the polypeptide, ϵN of a lysine at position Xaa (1) “1K”, the ϵN of a lysine at position Xaa (−1) “−1K”, or the ϵN of a lysine at position Xaa (−4) “−4K”.
  • Amino Acid Substitutions and Modifications
  • The polypeptides (e.g. lipidated polypeptides) of the invention may comprise one or more amino acid modifications or substitutions compared to the pramlintide sequence [SEQ ID NO: 1].
  • In some embodiments, the polypeptides (e.g. lipidated polypeptides) comprises one or more non-proteinogenic amino acids. Non-proteinogenic amino acids may include alpha methyl amino acids, D-enantiomers of naturally occurring amino acids, 2,4-diaminobutanoic acid (Dab), and (2S,4R)-4-hydroxypyrrolidine-2-carboxylic acid (Hyp). In some embodiments, the polypeptide (e.g. lipidated polypeptide) comprises one or more non-proteinogenic amino acids between positions 14-37, optionally at one or more of 14, 17 or 20-37.n some embodiments, the polypeptide (e.g. lipidated polypeptide) comprises one or more alpha methyl amino acids between positions 14-37, optionally at one or more of alpha methyl amino acids at positions 14, 17 or 20-37. Polypeptides (e.g. lipidated polypeptides) comprising one or more alpha methyl amino acids at positions 17, 21 or 23 are particularly preferred. Representative examples of alpha methyl amino acids include 2-amino-2-methylpropanoic acid (Aib), alpha-methyl glutamine (αMeGlu), alpha methyl phenylalanine (αMePhe or αMeF), alpha-methyl leucine (αMeLeu) and alpha-methyl serine (αMeSer). Thus, in certain embodiments, the alpha methyl amino acid can be Aib, αMeGlu, αMePhe, αMeLeu or αMeSer, or any combination thereof. In preferred embodiments, the polypeptide (e.g. lipidated polypeptide) comprises at least one alpha methyl amino acid, optionally selected from Aib, αMePhe and αMeSer. The reference to αMePhe and αMeF herein refers to (S)-2-amino-2-methyl-3-phenylpropanoic acid. The reference to αMeSer herein refers to (S)-2-amino-3-hydroxy-2-methylpropanoic acid. In preferred embodiments, the alpha methyl amino acid is Aib, αMePhe or αMeSer.
  • Figure US20220204580A1-20220630-C00011
  • In some embodiments, the polypeptide (e.g. lipidated polypeptide) comprises one or more non-proteinogenic amino acids between positions 14-37 selected from the group consisting of: 2,4-diaminobutanoic acid (Dab), (2S,4R)-4-hydroxypyrrolidine-2-carboxylic acid (Hyp), D-leucine (dL), D-isoleucine (dl) and D-proline (dP).
  • Figure US20220204580A1-20220630-C00012
  • In some embodiments, the polypeptide (e.g. lipidated polypeptide) does not comprise (2S)-2-aminohexanedioic acid) (Aad) and/or does not comprise Aad at positions 14-37.
  • In some embodiments, the polypeptide (e.g. lipidated polypeptide) does not comprise Aib at one or more of positions 15, 16, 17, 19 or 20. In alternative embodiments, the polypeptide (e.g. lipidated polypeptide) comprises Aib at one or more of positions 15, 16, 17, 19 or 20 and at least one different non-proteinogenic amino acid (e.g. an alpha methyl amino acid that is not Aib) at positions 14-37.
  • In some embodiments, the polypeptide (e.g. lipidated polypeptide) comprises one or more natural amino acid substitutions or modifications compared to the pramlintide sequence [SEQ ID NO: 1].
  • In preferred embodiments, the polypeptide (e.g. lipidated polypeptide) comprises one or more of the following natural amino acid substitutions or modifications: deleted 1K (A1K), Ile 4, Ala 4, Glu 14, His 14, Trp 15, Arg 17, Ser 17, Glu 17, Pro 20, Ile 20, His 21, Ala 21, Glu 21, Gly 21, Lys 21, Pro 21, Arg 21, Ser 21, His 22, Pro 24, Ala 25, Arg 26, Ser 28, His 31, Glu 31, Pro 31, Arg 31, His 34, Pro 33, Pro 34, Glu 35, Arg 35, Pro 35 and Pro 37.
  • It will be understood that the polypeptide (e.g. lipidated polypeptides) may comprise a combination of non-proteinogenic amino acids and natural amino acid substitutions or modifications compared to the pramlintide sequence [SEQ ID NO: 1].
  • In some aspects, there is provided a polypeptide (e.g. lipidated polypeptide) that is a pramlintide analogue, or a pharmaceutically acceptable salt thereof, comprising any of the amino acid sequence modification combinations set forth in Table 3.
  • TABLE 3
    Amino acid modifications to pramlintide sequence
    Sequence modification with respect to pramlintide
    −1G, −2G, 17Aib
    4A, 15W, 21P, 24P, 25A, 28S
    4I, 20I, 21A, 35R
    4I, 21Dab, 35R
    14Dab
    14Dab, 17Aib, 31E
    14Dab, 23αMePhe, 31E
    14Dab, 31E
    14E, 17Aib
    14E, 17Aib, 21H
    14E, 17R
    14E, 17R, 23αMePhe
    14E, 21Aib
    14H, 17Aib
    14H, 17Aib, 21Aib, 31E
    14H, 21Aib
    14H, 21Aib, 35E
    16dL, 21Aib
    17Aib
    17Aib, 37P
    17Aib, 21Aib
    17Aib, 21Aib, 37P
    17Aib, 21G
    17Aib, 21H
    17Aib, 21K
    17Aib, 21P
    17Aib, 21P, 31E
    17Aib, 21P, 35E
    17Aib, 21R
    17Aib, 21S
    17Aib, 21Dab
    17Aib, 21Dab, 31E
    17Aib, 22H
    17Aib, 22H, 35E
    17Aib, 23αMePhe
    17Aib, 26Aib
    17Aib, 26R
    17Aib, 27Aib
    17Aib, 27dL
    17Aib, 28Aib
    17Aib, 29Aib
    17Aib, 31Aib
    17Aib, 31E
    17Aib, 31H, 35E
    17Aib, 31P
    17Aib, 31R
    17Aib, 32Aib
    17Aib, 33Aib
    17Aib, 34Aib
    17Aib, 34H
    17Aib, 34P
    17Aib, 35Aib
    17Aib, 35E
    17Aib, 35R
    17E, 21Aib
    17R, 21Aib
    17R, 21Aib, 31Aib
    17R, 21Aib, 31E
    17R, 21Aib, 31R
    17R, 21Aib, 35Aib
    17R, 23αMePhe
    17R, 23αMePhe, 31E
    17R, 26Aib
    17S, 21Aib
    17S, 21Aib, 31H
    17S, 21Aib, 31P
    17S, 21Aib, 31R
    17S, 21Aib, 33P
    17S, 21Aib, 35P
    20αMeSer
    20P, 21P, 24P, 25A, 28S
    21Aib
    21Aib, 24P, 25A, 28S
    21Aib, 24P, 25A, 28S, 31Dab
    21Aib, 24P, 25A, 28S, 35Dab
    21Aib, 26dl
    21Aib, 26Aib
    21Aib, 27Aib
    21Aib, 27dL
    21Aib, 28Aib
    21Aib, 28dP
    21Aib, 31Aib
    21Aib, 31E
    21Aib, 31H
    21Aib, 31R
    21Aib, 33Aib
    21Aib, 34Aib
    21Aib, 35Aib
    21Aib, 35E
    21Aib, 35R
    21Aib, 36Aib
    21Aib, 37Aib
    21Aib, 37P
    21Aib, 24Hyp, 25A, 28S
    21Dab, 24P, 25A, 28S
    21Dab, 25Aib
    21Dab, 31E
    21P, 24P, 25A, 28S
    22Aib
    22H, 35E
    23αMePhe
    23αMePhe, 31E
    23aMePhe, 31R
    23αMePhe, 35R
    24Aib
    26Aib
    27Aib
    27dL
    28dP
    35R
    Δ1K, 41, 21Dab, 35R
    Δ1K, 14E, 17R, 23αMePhe
    Δ1K, 21Aib, 31R,
  • In one aspect, there is provided a polypeptide (e.g. lipidated polypeptide) that is a pramlintide analogue, or a pharmaceutically acceptable salt thereof, having an alpha methyl amino acid at position 23. In preferred embodiments, the alpha methyl amino acid is αMePhe.
  • In preferred embodiments of any aspect in which the polypeptide (e.g. lipidated polypeptide) comprises an alpha methyl amino acid (e.g. αMePhe) at position 23, the polypeptide (e.g. lipidated polypeptide) comprises any one of the following combinations of modifications:
      • 14E, 17R, 23αMePhe;
      • Δ1 K, 14E, 17R, 23αMePhe;
      • 14Dab, 23αMePhe, 31E;
      • 17Aib, 23αMePhe;
      • 17R, 23αMePhe, 31E;
      • 23αMePhe, 31E;
      • 23αMePhe, 31R, or
      • 23αMePhe, 35R.
  • In one aspect, there is provided a polypeptide (e.g. lipidated polypeptide) that is a pramlintide analogue, or a pharmaceutically acceptable salt thereof having at least two Aib residues. In preferred embodiments, the polypeptide (e.g. lipidated polypeptide) comprises Aib at at least two of positions 17 and 20-37. In particularly preferred embodiments, the polypeptide (e.g. lipidated polypeptide) comprises Aib at positions 21, 26, 27, 28, 29, 31, 32, 33, 34 and 35.
  • In preferred embodiments of any aspect in which the polypeptide (e.g. lipidated polypeptide) comprises at least two Aib residues, the polypeptide (e.g. lipidated polypeptide) comprises any one of the following combinations of modifications:
      • 14H, 17Aib, 21Aib, 31E;
      • 17Aib, 21Aib;
      • 17Aib, 21Aib, 37P;
      • 17Aib, 26Aib,
      • 17Aib, 27Aib;
      • 17Aib, 28Aib;
      • 17Aib, 29Aib;
      • 17Aib, 31Aib;
      • 17Aib, 32Aib;
      • 17Aib, 33Aib;
      • 17Aib, 34Aib;
      • 17Aib, 35Aib;
      • 17R, 21Aib, 31Aib;
      • 17R, 21Aib, 35Aib;
      • 21Aib, 26Aib;
      • 21Aib, 27Aib;
      • 21Aib, 28Aib;
      • 21Aib, 31Aib;
      • 21Aib, 33Aib;
      • 21Aib, 34Aib;
      • 21Aib, 35Aib;
      • 21Aib, 36Aib; or
      • 21Aib, 37Aib.
  • In one aspect, there is provided a polypeptide (e.g. lipidated polypeptide) that is a pramlintide analogue, or a pharmaceutically acceptable salt thereof, having an alpha methyl amino acid at position 21. In preferred embodiments, the alpha methyl amino acid is Aib.
  • In preferred embodiments of any aspect in which the polypeptide (e.g. lipidated polypeptide) comprises an alpha methyl amino acid (e.g. Aib) at position 21, the polypeptide (e.g. lipidated polypeptide) comprises any one of the following combinations of modifications:
      • 14E, 21Aib;
      • 14H, 17Aib, 21Aib, 31E;
      • 14H, 21Aib, 35E;
      • 14H, 21Aib;
      • 16dL, 21Aib;
      • 17Aib, 21Aib, 37P;
      • 17Aib, 21Aib;
      • 17E, 21Aib;
      • 17R, 21Aib, 31Aib;
      • 17R, 21Aib, 31E;
      • 17R, 21Aib, 31R,
      • 17R, 21Aib, 35Aib;
      • 17R, 21Aib;
      • 17S, 21Aib, 31H,
      • 17S, 21Aib, 31P,
      • 17S, 21Aib, 31R,
      • 17S, 21Aib, 33P,
      • 17S, 21Aib, 35P;
      • 17S, 21Aib;
      • 21Aib, 24Hyp, 25A, 28S
      • 21Aib, 24P, 25A, 28S, 31Dab,
      • 21Aib, 24P, 25A, 28S, 35Dab.
      • 21Aib, 24P, 25A, 28S;
      • 21Aib, 26Aib;
      • 21Aib, 26dl;
      • 21Aib, 27Aib;
      • 21Aib, 27dL;
      • 21Aib, 28Aib;
      • 21Aib, 28dP;
      • 21Aib, 31Aib;
      • 21Aib, 31E;
      • 21Aib, 31H,
      • 21Aib, 31R; Δ1K
      • 21Aib, 33Aib;
      • 21Aib, 34Aib;
      • 21Aib, 35Aib;
      • 21Aib, 35E;
      • 21Aib, 35R;
      • 21Aib, 36Aib;
      • 21Aib, 37Aib;
      • 21Aib, 37P; or
      • 21Aib.
  • In one aspect, there is provided a polypeptide (e.g. lipidated polypeptide) that is a pramlintide analogue, or a pharmaceutically acceptable salt thereof, having an alpha methyl amino acid at position 17. In preferred embodiments, the alpha methyl amino acid is Aib.
  • In preferred embodiments of any aspect in which the polypeptide (e.g. lipidated polypeptide) comprises an alpha methyl amino acid (e.g. Aib) at position 17, the polypeptide (e.g. lipidated polypeptide) comprises any one of the following combinations of modifications:
      • 14H, 17Aib;
      • −1G, −2G, 17Aib;
      • 17Aib, 23αMePhe;
      • 17Aib, 21Dab, 31E;
      • 17Aib, 21Dab,
      • 17Aib, 21Aib;
      • 14H, 17Aib, 21Aib, 31E;
      • 17Aib, 21Aib, 37P;
      • 17Aib, 26Aib;
      • 17Aib, 27Aib;
      • 17Aib, 28Aib;
      • 17Aib, 29Aib;
      • 17Aib, 31Aib;
      • 17Aib, 32Aib;
      • 17Aib, 33Aib;
      • 17Aib, 34Aib; or
      • 17Aib, 35Aib.
    Pharmacokinetics
  • The polypeptides (e.g. lipidated polypeptides) of the invention may exhibit favourable pharmacokinetic properties as compared to pramlintide. For example, the polypeptides (e.g. lipidated polypeptides) of the invention may have an extended half-life as compared to pramlintide.
  • As used herein, the term “half-life” is used to refer to the time taken for the concentration of isolated polypeptide in plasma to decline to 50% of its original level. Methods to determine the half-life of proteins are known in the art and are described in Example 4.
  • It will be recognised that an extended half-life is advantageous, as it permits the therapeutic proteins to be administered according to a safe and convenient dosing schedule, e.g. lower doses that can be administered less frequently. Moreover, the achievement of lower doses may provide further advantages such as the provision of an improved safety profile. To the contrary, pramlintide requires frequent and inconvenient administration.
  • The present inventors have shown that the polypeptides (e.g. lipidated polypeptides) of the invention may have a half-life of at least 4 hours in rat models (see Example 4). In embodiments, the polypeptide (e.g. lipidated polypeptide) has a half-life of at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 5 hours, at least 6 hours, at least 7 hours, at least 8 hours, at least 9 hours, at least 10 hours, at least 11 hours, at least 12 hours, at least 13 hours or at least 14 hours in rat models. In preferred embodiments, the polypeptide (e.g. lipidated polypeptide) has a half-life of at least 14 hours.
  • Reduced Fibrillation
  • The polypeptides (e.g. lipidated polypeptides) of the invention may exhibit reduced tendency to undergo fibrillation in pharmaceutically relevant aqueous media, especially at pH values in the range from 4 to 7, as compared to lipidated pramlintide. In some embodiments, the polypeptide (e.g. lipidated polypeptide) exhibits reduced tendency to undergo fibrillation in pharmaceutically relevant aqueous media, especially at pH values in the range from 4 to 7, as compared to pramlintide which is lipidated in a similar manner e.g. the same lipid is attached, the lipid is attached through the same linker and/or the lipid is attached at the same position. Exemplary lipidated pramlintide molecules are given in Table 1, for example SEQ ID NO. 3, 4, 5, 6, 7, 112 and 113.
  • Accordingly, the polypeptides (e.g. lipidated polypeptides) of the invention may be suited for formulation in acidic media (e.g. pH 4) and in neutral or near-neutral media (e.g. pH 7 or 7.4). Such polypeptides (e.g. lipidated polypeptides) may be well suited for co-formulation with, for example, insulin, various insulin analogues and/or other therapeutic (e.g. anti-diabetic or anti-obesity) agents that require a neutral or near-neutral formulation pH.
  • In some embodiments, the polypeptide (e.g. lipidated polypeptide) shows no detectable fibrillation after about 5 hours, after about 7 hours, after about 9 hours, after about 11 hours, after about 13 hours, after about 15 hours, after about 17 hours or after about 20 hours at pH 4 and 37° C., e.g. under the conditions described in Example 3.
  • In preferred embodiments, the polypeptide (e.g. lipidated polypeptide) shows no detectable fibrillation after about 48 hours, after about 72 hours, after about 96 hours, after about 108 hours, after about 120 hours, after 132 about hours or after about 144 hours at pH 4 and 37° C., e.g. under the conditions described in Example 3. In particularly preferred embodiments, the polypeptide (e.g. lipidated polypeptide) shows no detectable fibrillation after 144 hours at pH 4 and 37° C., e.g. under the conditions described in Example 3.
  • In some embodiments, the formation of fibrils is detected by an increase in fluorescence intensity in a Thioflavin T fibrillation assay, e.g. as described in Example 3.
  • In preferred embodiments, the polypeptides (e.g. lipidated polypeptides) of the invention are soluble at concentrations required for therapeutic efficacy. In some embodiments, the lipidated polypeptides of the invention are soluble at a concentration of at least 1 mg/mL under the conditions described in Example 3.
  • Efficacy
  • The polypeptides (e.g. lipidated polypeptides) of the invention are amylin receptor agonists, i.e. they are capable of binding to, and inducing signalling by, one or more receptors or receptor complexes regarded as physiological receptors for human amylin. These include the human calcitonin receptor hCTR, as well as complexes comprising the human calcitonin receptor hCTR and at least one of the human receptor activity modifying proteins designated hRAMP1, hRAMP2 and hRAMP3. Complexes between hCTR and hRAMP1, hRAMP2 and hRAMP3 are designated hAMYR1, hAMYR2 and hAMYR3 (i.e. human amylin receptors 1, 2 and 3) respectively. In some embodiments, a compound is considered an amylin receptor agonist if it has agonist activity at one or more of hAMYR1, hAMYR2 and hAMYR3. For example, a compound may be considered an amylin receptor agonist if it has agonist activity at hAMYR3.
  • The ability to induce cAMP formation as a result of binding to the relevant receptor or receptor complex is typically regarded as indicative of agonist activity. Other intracellular signaling pathways or events may also be used as readouts for amylin receptor agonist activity. These may include calcium release, arrestin recruitment, receptor internalization, kinase activation or inactivation, lipase activation, inositol phosphate release, diacylglycerol release or nuclear transcription factor translocation.
  • EC50 values may be used as a measure of agonist potency at a given receptor. An EC50 value is a measure of the concentration of a compound required to achieve half of that compound's maximal activity in a particular assay, for example a cAMP assay as described in Example 2. In Example 2, the present inventors have shown that certain polypeptides (e.g. lipidated polypeptides) disclosed herein exhibit greater or similar selectivity to hAMYR over hCTR as pramlintide, as measured using cAMP release from binding to hAMYR and hCTR. Pramlintide exhibits at least 10-fold selectivity to hAMYR as compared to hCTR.
  • The polypeptides (e.g. lipidated polypeptides) of the invention may exhibit improved efficacy, e.g. as amylin receptor agonists, as compared to lipidated pramlintide.
  • In some embodiments, the polypeptide (e.g. lipidated polypeptide) has at least about 1-fold selectivity to hAMYR over hCTR, optionally at least about 2-fold, at least about 4-fold, at least about 6-fold, at least about 8-fold, at least about 10-fold, at least about 12-fold, at least about 14-fold, at least about 16-fold, at least about 18-fold, at least about 20-fold, at least about 50-fold, at least about 75-fold, or at least about 100-fold selectivity to hAMYR over hCTR. In preferred embodiments, the polypeptide (e.g. lipidated polypeptide) has at least about 10-fold selectivity to hAMYR over hCTR.
  • In some embodiments, the polypeptide (e.g. lipidated polypeptide) has around 12-20 fold, around 14-18 fold, optionally around 16-fold selectivity to hAMYR over hCTR.
  • n some embodiments, the isolated polypeptide has an EC50 measured under the conditions described in Example 2 (i.e. containing 0.1% bovine serum albumin (BSA)) of below about 1.4 nM, below about 1.2 nM, below about 1 nM, below about 0.8 nM, below about 0.6 nM, below about 0.4nM, below about 0.3 nM, or below about 0.2 nM.
  • Chemical Stability
  • The polypeptides (e.g. lipidated polypeptides) of the invention may be chemically stable, e.g. they may form in a formulation an acceptable percentage of degradation products produced over a defined period of time by chemical pathways, such as deamidation, aggregation, or oxidation.
  • The polypeptides (e.g. lipidated polypeptides) of the invention may be chemically conjugated to a protein or polymeric drug carrier, or formulated in an advance drug delivery system, that enhances the chemical stability and/or physical stability and/or the circulatory exposure of the polypeptide.
  • In some aspects, there is provided a polypeptide or a pharmaceutically acceptable salt thereof, wherein the polypeptide comprises any one of the lipid linkers as set forth in Table 2 and any one of the sequence modifications as set forth in Table 3.
  • In some aspects, there is provided a polypeptide or a pharmaceutically acceptable salt thereof, wherein the polypeptide comprises the lipid linker and amino acid sequence modification combinations set forth in Table 4.
  • TABLE 4
    Lipidated polypeptides
    Acylation Sequence modification with
    ID Lipid Linker site respect to pram lintide
    8 C18diacid γE-γE   1K 21Dab, 24P, 25A, 28S
    9 C18diacid γE-γE   1K 21Aib, 24P, 25A, 28S
    10 C18diacid γE N- 14E, 17R, 23αMePhe, and Δ1K
    terminal
    11 C18diacid γE-γE   1K 14E, 17R, 23αMePhe
    12 C18diacid γE −1K 14E, 17R, 23αMePhe
    13 C18diacid γE-γE   1K 23αMePhe
    14 C18diacid γE −1K 23αMePhe
    15 C18diacid γE −1K 17R, 23αMePhe, 31E
    16 C18diacid γE   1K 17R, 23αMePhe, 31E
    17 C18diacid γE-(O2Oc)-   1K 17R, 23αMePhe, 31E
    (O2Oc)
    18 C18diacid γE-γE   1K 17R, 23αMePhe, 31E
    19 C18diacid γE   1K 17R, 23αMePhe
    20 C18diacid γE-γE   1K 17R, 23αMePhe
    21 C18diacid γE-γE   1K 23αMePhe, 35R
    22 C18diacid γE-γE   1K 20αMeS
    23 C18diacid γE-γE   1K 23αMePhe, 31E
    24 C18diacid γE-γE   1K 23αMePhe, 31R
    25 C18diacid γE   1K 23αMePhe, 31R
    26 C18diacid γE-γE   1K 17Aib, 23αMePhe
    27 C18diacid γE-γE   1K 41, 21Dab, 35R
    28 C18diacid γE −1K 21Dab, 31E
    29 C18diacid γE −1K 21Dab
    30 C18diacid γE −1K 17Aib, 21Dab, 31E
    31 C18diacid γE −1K 21Dab, 25Aib
    32 C18diacid γE −1K 14Dab, 23αMePhe, 31E
    33 C18diacid γE −1K 14Dab
    34 C18diacid γE −1K 14Dab, 31E
    35 C18diacid γE −1K 21Aib
    36 C18diacid γE −1K 17Aib, 21Aib
    37 C18diacid γE −1K 17S, 21Aib
    38 C18diacid γE −1K 14E, 21Aib
    39 C18diacid γE −1K 17E, 21Aib
    40 C18diacid γE −1K 21Aib, 31H
    41 C18diacid γE −1K 21Aib, 31E
    42 C18diacid γE −1K 21Aib, 35E
    43 C18diacid γE −1K 17R, 21Aib, 31E
    44 C18diacid γE   1K 17R, 21Aib, 31E
    45 C18diacid γE-(O2Oc)-   1K 17R, 21Aib, 31E
    (O2Oc)
    46 C18diacid γE −1K 14H, 21Aib
    47 C18diacid γE −1K 14H, 21Aib, 35E
    48 C18diacid γE   1K 17R, 21Aib
    49 C18diacid γE-γE   1K 21Aib, 31E
    50 C18diacid γE-γE   1K 17R, 21Aib, 31E
    51 C18diacid γE −1K 17S, 21Aib, 31H
    52 C18diacid γE −1K 17S, 21Aib, 31R
    53 C18diacid γE −1K 17S, 21Aib, 31P
    54 C18diacid γE −1K 17S, 21Aib, 33P
    55 C18diacid γE −1K 17S, 21Aib, 35P
    56 C18diacid γE   1K 21Aib
    58 C18diacid γE −1K 21Aib, 37P
    59 C18diacid γE   1K 21Aib, 27dL
    60 C18diacid γE   1K 21Aib, 28dP
    61 C18diacid γE   1K 21Aib, 26dl
    62 C18diacid γE   1K 16dL, 21Aib
    63 C18diacid γE   1K 21Aib, 31R
    64 C18diacid γE   1K 21Aib, 35R
    65 C18diacid γE −1K 17R, 21Aib
    66 C18diacid γE-γE −1K 17R, 21Aib
    67 C18diacid γE-γE   1K 21Aib, 31R
    68 C18diacid γE N- 21Aib, 31R, Δ1K
    terminal
    69 C18diacid γE −1K 21Aib, 31R
    70 C18diacid γE-γE −1K 21Aib, 31R
    71 C18diacid γE −1K 21Aib, 31R
    72 C18diacid γE-γE   1K 21Aib, 35R
    73 C18diacid γE −1K 21Aib, 35R
    74 C18diacid γE-γE −1K 21Aib, 35R
    75 C18diacid γE-γE   1K 17R, 21Aib
    76 C18diacid γE   1K 17R, 21Aib, 31R
    77 C18diacid γE-γE   1K 17R, 21Aib, 31R
    78 C18diacid γE −1K 21Aib, 26Aib
    79 C18diacid γE −1K 21Aib, 27Aib
    80 C18diacid γE −1K 21Aib, 31Aib
    81 C18diacid γE −1K 21Aib, 33Aib
    82 C18diacid γE −1K 21Aib, 35Aib
    83 C18diacid γE −1K 21Aib, 36Aib
    84 C18diacid γE −1K 21Aib, 34Aib
    85 C18diacid γE −1K 21Aib, 37Aib
    86 C18diacid γE −1K 14H, 17Aib, 21Aib, 31E
    87 C18diacid γE −1K 17Aib, 21Aib, 37P
    88 C18diacid γE −1K 21Aib, 28Aib
    89 C18diacid γE-γE   1K 21Aib, 31Aib
    90 C18diacid γE-γE   1K 17R, 21Aib, 31Aib
    91 C18diacid γE-γE   1K 21Aib, 35Aib
    92 C18diacid γE-γE   1K 17R, 21Aib, 35Aib
    94 C18diacid γE −1K 17Aib, 26Aib
    95 C18diacid γE −1K 17Aib, 27Aib
    96 C18diacid γE −1K 17Aib, 28Aib
    97 C18diacid γE −1K 17Aib, 29Aib
    98 C18diacid γE −1K 17Aib, 31Aib
    99 C18diacid γE −1K 17Aib, 32Aib
    100 C18diacid γE −1K 17Aib, 33Aib
    101 C18diacid γE −1K 17Aib, 34Aib
    102 C18diacid γE −1K 17Aib, 35Aib
    103 C18diacid γE   1K 27dL
    104 C18diacid γE   1K 28dP
    106 C18diacid γE −1K 26Aib
    107 C18diacid γE −1K 17R, 26Aib
    108 C18diacid γE −1K 27Aib
    109 C18diacid γE −1K 22Aib
    110 C18diacid γE −1K 24Aib
    111 C18diacid γE −1K 22H, 35E
    114 C20diacid γE-γE  1K 35R
    115 C20diacid γE-γE  1K 21P, 24P, 25A, 28S
    116 C20diacid γE-γE  1K 14E, 17R
    117 C20diacid γE-γE  1K 21Aib, 24P, 25A, 28S
    118 C20diacid γE-γE  1K 4I, 201, 21A, 35R
    119 C20diacid γE-γE  1K 20P, 21P, 24P, 25A, 28S
    120 C20diacid γE-γE  1K 4A, 15W, 21P, 24P, 25A, 28S
    121 C20diacid γE-γE  1K 21Dab, 24Hyp, 25A, 28S
    122 C18diacid γE-γE  1K 21Aib, 24P, 25A, 28S, 31Dab
    123 C20diacid γE-γE  1K 21Aib, 24P, 25A, 28S, 31Dab
    124 C18diacid γE-γE  1K 21Aib, 24P, 25A, 28S, 35Dab
    125 C20diacid γE-γE  1K 21Aib, 24P, 25A, 28S, 35Dab
    126 C18diacid γE −1K 17Aib, 21Dab
    127 C18diacid γE-γE   1K 17Aib
    128 C18diacid γE −1K 14E, 17Aib
    129 C18diacid γE −1K 17Aib
    130 C18diacid γE −1K 14E, 17Aib, 21H
    131 C18diacid γE −1K 17Aib, 21H
    132 C18diacid γE −1K 17Aib, 21P
    133 C18diacid γE −1K 17Aib, 21S
    134 C18diacid γE −1K 17Aib, 31P
    135 C18diacid γE −1K 17Aib, 22H
    136 C18diacid γE −1K 17Aib, 37P
    137 C18diacid γE −1K 17Aib, 21R
    138 C18diacid γE −1K 17Aib, 21P, 31E
    139 C18diacid γE −1K 17Aib, 21P, 35E
    140 C18diacid γE −1K 17Aib, 31E
    141 C18diacid γE −1K 17Aib, 35R
    142 C18diacid γE −1K 17Aib, 35E
    143 C18diacid γE −3K −1G, −2G, 17Aib
    144 C18diacid γE   1K 14H, 17Aib
    145 C18diacid γE −1K 17Aib, 34H
    146 C18diacid γE −1K 17Aib, 31H, 35E
    147 C18diacid γE −1K 17Aib, 22H, 35E
    148 C18diacid γE −1K 17Aib, 34P
    149 C18diacid γE   1K 17Aib
    150 C18diacid γE   1K 17Aib, 21Dab
    151 C18diacid γE   1K 17Aib, 27dL
    152 C18diacid γE −1K 17Aib, 26R
    153 C18diacid γE −1K 17Aib, 21K
    154 C18diacid γE −1K 17Aib, 21G
    155 C18diacid γE   1K 17Aib, 31R
    156 C18diacid γE −1K 14Dab, 17Aib, 31E
    157 C18diacid Nil N- 21Aib, 31Aib
    terminal
    158 C18diacid Nil   1K 21Aib, 31Aib
  • Process
  • The polypeptides (e.g. lipidated polypeptides) of the invention may be produced by any method known in the art. The production of polypeptides such as amylin or analogues thereof is well known in the art. The polypeptide (e.g. lipidated polypeptides) of the invention can thus be produced by chemical synthesis, e.g. solid phase polypeptide synthesis using t-Boc or Fmoc chemistry, or other well-established techniques. They may alternatively be produced by recombinant expression of a nucleic acid molecule encoding a fusion polypeptide in a host cell. Following synthesis, the polypeptides (e.g. lipidated polypeptides) of the invention may optionally be isolated or purified.
  • Therapeutic Methods
  • In further aspects, the polypeptides (e.g. lipidated polypeptides) of the invention are provided in a pharmaceutical composition.
  • The pharmaceutical compositions of the invention may comprise one or more excipient(s). Pharmaceutically acceptable excipients are known in the art, see for instance Remington's Pharmaceutical Sciences (by Joseph P. Remington, 18th ed., Mack Publishing Co., Easton, Pa.), which is incorporated herein in its entirety.
  • The present invention encompasses therapies which involve administering the polypeptides (e.g. lipidated polypeptides) of the invention to an animal, in particular a mammal, for instance a human, for preventing, treating, or ameliorating symptoms associated with a disease, disorder, or infection.
  • Accordingly, the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention may be used in therapy, for example for treating a disease or disorder. Also provided is a method of treating a disease or disorder comprising administering to a subject or patient in need thereof a therapeutically effective amount of the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention. The use or method may comprise administering a therapeutically effective schedule that has less frequent doses of the polypeptides (e.g. lipidated polypeptides) of the invention than the therapeutically effective dosing schedule of pramlintide.
  • It will be understood that the polypeptides (e.g. lipidated polypeptides) of the invention may be used in the treatment and/or prevention of obesity, metabolic diseases such as diabetes (e.g. type 1 or type 2 diabetes), and/or obesity-related conditions.
  • Accordingly, the polypeptides (e.g. lipidated polypeptides) of the invention may be used in a method of treating obesity, overweight, morbid obesity, obesity prior to surgery, obesity-linked inflammation, obesity-linked gallbladder disease, sleep apnea and respiratory problems, hyperlipidemia, degeneration of cartilage, osteoarthritis, or reproductive health complications of obesity or overweight such as infertility in a subject, the method comprising administering a therapeutically effective amount of the polypeptide (e.g. lipidated polypeptide) to the subject.
  • This is also provided a method of inhibiting or reducing weight gain, promoting weight loss, reducing food intake, and/or reducing excess body weight, the method comprising administering the polypeptide (e.g. lipidated polypeptide) of the invention to the subject.
  • Metabolic diseases that may be treated by the polypeptide (e.g. lipidated polypeptide) of the invention include diabetes, type 1 diabetes, type 2 diabetes, gestational diabetes, pre-diabetes, insulin resistance, impaired glucose tolerance (IGI), disease states associated with elevated blood glucose levels, metabolic disease including metabolic syndrome, or hyperglycemia e.g. abnormal postprandial hyperglycemia.
  • In preferred embodiments, the polypeptides (e.g. lipidated polypeptides) of the invention are used for the treatment of type 1 diabetes or type 2 diabetes.
  • The polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention may be used for treating, inhibiting or reducing weight gain, promoting weight loss, reducing food intake, and/or reducing excess body weight.
  • The polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention may be used in the treatment and/or prevention of an eating disorder, Alzheimer's disease, hepatic steatosis (“fatty liver”), kidney failure, arteriosclerosis (e.g. atherosclerosis), cardiovascular disease, macrovascular disease, microvascular disease, diabetic heart (including diabetic cardiomyopathy and heart failure as a diabetic complication), coronary heart disease, peripheral artery disease or stroke, cancer, dumping syndrome, hypertension e.g. pulmonary hypertension, or dyslipidemia e.g. atherogenic dyslipidemia, cholescystitis, or short bowel syndrome.
  • The route of administration of polypeptides (e.g. lipidated polypeptides) of the invention, or pharmaceutical compositions thereof, can be, for example, oral, parenteral, by inhalation or topical. In preferred embodiments, the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition thereof is administered by parenteral administration to a subject or patient. The term “parenteral” as used herein includes, e.g., intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, rectal, or vaginal administration. In preferred embodiments, the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition thereof is administered by injection, such as by intravenous, subcutaneous or intramuscular injection, to a subject or patient. In particularly preferred embodiments, the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition thereof is administered by subcutaneous injection. Administration by injection, such as by subcutaneous injection, offers the advantage of better comfort for the subject or patient and the opportunity to administer to a subject or patient outside of a hospital setting. In some embodiments, the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition thereof is administered by self-administration.
  • In some embodiments the subject or patient is a mammal, in particular a human.
  • In some embodiments, the polypeptide or pharmaceutical composition is administered to the subject in combination with insulin.
  • Articles of Manufacture and Kits
  • In other aspects, the present invention provides an article of manufacture comprising the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention.
  • In yet other aspects, the present invention provides a kit comprising the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention. The kit may comprise a package containing the polypeptide (e.g. lipidated polypeptide) or pharmaceutical composition, optionally with instructions. In some embodiments, the polypeptides (e.g. lipidated polypeptides) or pharmaceutical compositions of the invention are formulated in single dose vials or a container closure system (e.g. pre-filled syringe). Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Singleton, et al., DICTIONARY OF MICROBIOLOGY AND MOLECULAR BIOLOGY, 20 ED., John Wiley and Sons, New York (1994), and Hale & Marham, THE HARPER COLLINS DICTIONARY OF BIOLOGY, Harper Perennial, NY (1991) provide the skilled person with a general dictionary of many of the terms used in this disclosure.
  • This disclosure is not limited by the exemplary methods and materials disclosed herein, and any methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of this disclosure.
  • Unless otherwise indicated, any nucleic acid sequences are written left to right in 5′ to 3′ orientation; amino acid sequences are written left to right in amino to carboxy orientation, respectively.
  • It must be noted that as used herein and in the appended claims, the singular forms “a”, “an”, and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “an agent” includes a plurality of such agents and reference to “the agent” includes reference to one or more agents and equivalents thereof known to those skilled in the art, and so forth.
  • “About” may generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, and more typically, within 5% of a given value or range of values. Optionally, the term “about” shall be understood herein as plus or minus (±) 5%, optionally ±4%, ±3%, ±2%, ±1%, ±0.5%, ±0.1%, of the numerical value of the number with which it is being used.
  • Embodiments described herein as “comprising” one or more features may also be considered as disclosure of the corresponding embodiments “consisting of” such features.
  • The term “pharmaceutically acceptable” as used herein means approved by a regulatory agency of the Federal or a state government, or listed in the U.S. Pharmacopeia, European Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • Concentrations, amounts, volumes, percentages and other numerical values may be presented herein in a range format. It is also to be understood that such range format is used merely for convenience and brevity and should be interpreted flexibly to include not only the numerical values explicitly recited as the limits of the range but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited.
  • The above embodiments are to be understood as illustrative examples. Further embodiments are envisaged. It is to be understood that any feature described in relation to any one embodiment may be used alone, or in combination with other features described, and may also be used in combination with one or more features of any other of the embodiments, or any combination of any other of the embodiments. Furthermore, equivalents and modifications not described above may also be employed without departing from the scope of the invention, which is defined in the accompanying claims.
  • In the context of the present disclosure other examples and variations of the polypeptides (e.g. lipidated polypeptides) and methods described herein will be apparent to a person of skill in the art.
  • Other examples and variations are within the scope of the disclosure, as set out in the appended claims.
  • All documents cited herein are each entirely incorporated by reference herein, including all data, tables, figures, and text presented in the cited documents.
  • EXAMPLES Example 1 Generation of lipidated pramlintide Analogue Peptides
  • Lipidated pramlintide analogue peptides were synthesized as C-terminal carboxamides using rink amide MBHA resin (100-200 mesh). All peptides were prepared by automated synthesis using a Liberty Blue™ microwave solid phase peptide synthesizer (CEM Corporation, N.C., USA) using the Fmoc/tBu protocol. Manufacturer-supplied protocols were applied for coupling of amino acids in DMF and deprotection of Fmoc protecting group using piperidine in DMF (20% v/v). Asparagine, cysteine, glutamine and histidine were incorporated as their sidechain trityl (Trt) derivatives. Lysine was incorporated as the sidechain tert-butyloxycarbonyl (Boc) derivative. Serine, threonine and tyrosine were incorporated as sidechain tert-butyl (tBu) ethers, and aspartate and glutamate as their sidechain OtBu esters. Arginine was incorporated as the sidechain 2,2,4,6,7-pentamethyldihydrobenzofuran-5-sulfonyl (Pbf) derivative.
  • Boc-Lys(Fmoc) was incorporated when a subsequent chemical modification of the N-terminal lysine side chain was required. Upon completion of the peptide chain elongation, coupling of an albumin binding moiety, such as a lipid, was performed manually using HATU as a coupling reagent in the presence of DI PEA.
  • Peptides were cleaved from the solid support by treatment with a mixture of TFA:TIS:EDT:thioanisole:water (90:2.5:2.5:2.5:2.5 v/v) for 4 h with agitation at room temperature. Thereafter, the cleavage mixtures were filtered, concentrated in vacuo, precipitated and washed with diethyl ether and solids were isolated by centrifugation. The linear crude peptides were dried under a flow of nitrogen and dissolved in 20% MeCN in water (v/v) with 1% TFA (v/v) and filtered. The crude linear peptides were purified using a preparative RP-HPLC on a Varian SD-1 Prep Star binary pump system, monitoring by UV absorption at 210 nm using an Xbridge C18-A stationary phase (19.0×250 mm, 5 micron) column eluting a linear solvent gradient of 25-70% MeCN (0.1% TFA v/v) in water (0.1% TFA v/v) over 25 min.
  • The linear purified peptides were cyclised by treatment with iodine (1% w/v in methanol) for 10 min at room temperature and excess iodine was reduced by treatment with ascorbic acid (1% w/v in water). The cyclic crude peptides were re-purified as described above. The purified fractions were pooled, frozen and lyophilised.
  • LC/MS characterisation of purified peptides were performed on a Waters MassLynx 3100 platform using a XBridge C18 stationary phase (4.6×100 mm, 3 micron) eluting a linear binary gradient of 10-90% MeCN (0.1% TFA v/v) in water (0.1% TFA v/v) over 10 minutes at 1.5 mL/min at ambient temperature. Analytes were detected by both UV absorption at 210 nm and ionization using a Waters 3100 mass detector (ESI+ mode). Analytical RP-HPLC characterisation was performed on an Agilent 1260 Infinity system using an Agilent Polaris C8-A stationary phase (4.6×100 mm, 3 micron) eluting a linear binary gradient of 10-90% MeCN (0.1% TFA v/v) in water (0.1% TFA v/v) at 1.5 mL/min over 15 minutes at 40° C.
  • Example 2 In Vitro Potency of lipidated pramlintide Analogue Peptides in Human or Rat amylin or Calcitonin Receptor Cells
  • The functional activities of lipidated pramlintide analogue peptides, such as cAMP production, were tested in 1321N1 cell line with stable recombinant expression of human calcitonin receptor (hCTR) or human amylin receptor (calcitonin receptor co-expressed with receptor activity modifying protein, RAMP3) (hAMYR3) or HEK cells with stable recombinant expression of rat calcitonin receptor (rat CTR) or rat amylin receptor (calcitonin receptor co-expressed with receptor activity modifying protein, RAM P3) (Rat AMYR3).
  • Cryopreserved cell stock was thawed rapidly in a water-bath, suspended in assay buffer (0.1% BSA (Sigma #A3059) in HBSS (Sigma #H8264) with 25 mM HEPES, pH 7.4 and containing 0.5 mM IB MX (Sigma#17018)) and spun at 240×g for 5 minutes. Cells were re-suspended in assay buffer at a batch-dependent optimized concentration (e.g. hCTR cells at 0.125×105 cells/mL, hAMYR3 cells at 0.125×105 cells/mL, rat CTR cells at at 1×105 cells/mL, rat AMYR3 at 2×105 cells/mL).
  • The test peptide stock was prepared in DMSO and diluted in assay buffer to reach stated concentrations and transferred in duplicates into a 384-black shallow well microtitre assay plate (Corning #3676). Cells were added to the assay plate, incubated at room temperature for 30 minutes and the cAMP level measured using cAMP dynamic 2 HTRF kit (Cisbio, Cat #62AM4PEJ), following the two step protocol as per manufacturer's recommendations. The plates were read on an Envision (Perkin Elmer) using excitation wavelength of 320 nm and emission wavelengths of 620 nm & 665 nm.
  • Data was transformed to % Delta F as described in the manufacturer's guidelines and analyzed as percent activation of maximal amylin or calcitonin effect by 4-parameter logistic fit to determine EC50 values. The selectivity of a peptide to hAMYR vs hCTR is defined as a ratio of EC50 values at the two receptors.
  • All tested compounds show measurable potency in hAMYR and hCTR. Analogues that show >10 fold selectivity for hAMYR over hCTR are preferred.
  • TABLE 5
    in vitro potency of lipidated pramlintide analogues at
    human amylin3 and calcitonin receptors
    EC50 (pM)
    Peptide hAMYR3 hCTR Ratio
    1 10 160 16
    3 246 1353 5
    4 248 7532 30
    5 174 3605 21
    6 68 310 5
    7 173 2159 12
    8 375 20228 54
    9 320 13521 42
    10 183 2728 15
    11 177 3640 21
    12 158 1224 8
    13 196 2043 10
    14 220 1350 6
    15 522 5461 10
    16 221 2018 9
    17 365 7083 19
    18 319 4686 15
    19 278 4041 15
    20 174 4593 26
    21 136 3408 25
    22 162 6539 40
    23 238 589 2
    24 127 1857 15
    25 105 1248 12
    26 409 2797 7
    27 134 731 5
    28 390 9142 23
    29 243 8211 34
    30 873 10549 12
    31 349 1280 4
    32 742 11149 15
    33 598 15830 26
    34 877 17733 20
    35 259 14153 55
    36 331 6394 19
    37 249 1962 8
    38 190 1049 6
    39 1048 10362 10
    40 228 3329 15
    41 178 1184 7
    42 470 2954 6
    43 647 19046 29
    44 284 6262 22
    45 588 16891 29
    46 109 362 3
    47 218 468 2
    48 126 5273 42
    49 334 4142 12
    50 349 18927 54
    51 126 1645 13
    52 89 1104 12
    53 98 235 2
    54 7587 1507 0.2
    55 678 1874 3
    56 70 493 7
    58 71 65 1
    59 157 809 5
    60 96 1027 11
    61 99 654 7
    62 167 7249 43
    63 76 375 5
    64 149 1278 9
    65 207 6400 31
    66 418 7011 17
    67 132 4006 30
    68 192 1774 9
    69 164 1572 10
    70 235 5405 23
    71 211 1385 7
    72 227 4004 18
    73 256 1821 7
    74 356 4574 13
    75 161 15998 99
    76 209 22291 107
    77 231 2520 11
    78 288 3147 11
    79 659 4799 7
    80 407 2970 7
    81 404 2764 7
    82 514 4277 8
    83 599 6342 11
    84 118 401 3
    85 806 3104 4
    86 119 311 3
    87 71 74 1
    88 281 1172 4
    89 447 8239 18
    90 1056 15595 15
    91 200 2357 12
    92 341 44414 130
    94 322 1903 6
    95 691 1172 2
    96 556 859 2
    97 684 944 1
    98 942 972 1
    99 3798 1825 0.5
    100 364 459 1
    101 177 441 2
    102 479 1630 3
    103 161 1881 12
    104 79 1816 23
    106 473 3420 7
    107 1071 26264 25
    108 801 6808 8
    109 82 176 2
    110 204 541 3
    111 403 1875 5
    112 861 1335 2
    113 1173 18495 16
    114 1317 15796 12
    115 703 7645 11
    116 1207 9557 8
    117 770 3190 4
    118 417 792 2
    119 1461 34657 24
    120 1769 10717 6
    121 1328 6074 5
    122 655 6181 9
    123 873 4793 5
    124 1129 6633 6
    125 1201 10197 8
    126 445 8453 19
    127 364 7127 19.6
    128 118 568 4.8
    129 553 5750 10.4
    130 446 6686 15
    131 737 15143 20.5
    132 416 3848 9.3
    133 378 2578 6.8
    134 281 2223 7.9
    135 296 345 1.2
    136 122 101 0.8
    137 238 1504 6.3
    138 612 2506 4.1
    139 285 1043 3.7
    140 656 6046 9.2
    141 414 3950 9.5
    142 730 2270 3.1
    143 592 6937 11.7
    144 138 725 5.3
    145 417 1876 4.5
    146 2533 4874 1.9
    147 802 1508 1.9
    148 191 728 3.8
    149 130 1200 9.2
    150 179 4181 23.4
    151 61 898 14.7
    154 574 1540 2.7
    155 329 3930 11.9
    156 3749 83771 22.3
  • TABLE 6
    in vitro potency of lipidated pramlintide analogues
    at rat amylin3 and calcitonin receptors
    EC50 (pM)
    Peptide Rat AMYR3 Rat CTR
    1 0.4 70.0
    3 2.9 186.0
    8 25.6 19856.1
    9 17.8 9724.4
    10 12.1 2781.5
    12 5.0 106.8
    15 10.3 1821.8
    18 16.5 3086.9
    20 10.4 1854.2
    24 17.7 754.9
    35 5.9 514.1
    38 5.1 20.3
    40 12.0 782.1
    41 8.2 924.8
    43 14.5 3650.9
    44 18.5 2237.3
    48 4.8 2431.0
    112 16.0 258.5
    113 32.6 10375.7
    114 12.2 4873.7
    115 22.6 22552.0
    116 23.7 9052.7
    129 11.2 2869.2
    140 19.5 1218.4
    149 5.6 521.8
  • Example 3 Thioflavin T Fibrillation Assay
  • Peptide aggregation that form fibrils is an indication of physical instability. Fibril formation in solution poses a significant risk for the stability of injectable peptide drug products. Thioflavin T (ThT) fibrillation assay is a useful tool to assess the aggregation kinetics of a peptide or protein under accelerated and stressed conditions that can be used to forecast the long-term viability of a compound in solution.
  • ThT can selectively bind amyloid fibrils and the resultant complex emits strong fluorescence signal at 482 nm when excited at 450 nm (Anal Biochem. 1989 March; 177(2):244-9). Monitoring of the change in fluorescence signal is an established method to study the fibril forming potential of peptides and proteins.
  • ThT (purchased from Sigma Aldrich) stock solution is prepared by dissolving the ThT powder in Milli-Q water and filtered to obtain a 0.25 mM solution. The concentration of the solution is measured at 412 nm using an extinction coefficient of 36 mM−1 cm−1. Test peptides were dissolved at 1 mg/mL in 25 mM sodium acetate buffer pH 4.0.
  • 100 μL aliquot of peptide solution and 5 μL aliquot of ThT solution were placed in a clear bottom black fluorescence 96-well plate. 5 replicates of each test samples were placed in the same row of the plate. Buffer was placed in control wells for baseline correction. All empty wells were filled with water to prevent evaporation. The plate was sealed with aluminium seal and placed in fluorescence plate reader and incubated for 6 days at 37° C. with intermittent orbital shaking at 500 to 750 rpm. The fluorescence intensity was measured every 30 min using excitation at 444 nm and emission at 480 nm.
  • The fibril forming potential of the test peptides was determined by measuring the average time taken to detect an increase in baseline corrected fluorescence intensity. A time>144 h indicates no increase in fluorescence intensity, relative to baseline, during the course of the experiment.
  • Conjugating pramlintide to a lipid (for example, as in SEQ ID NO. 3, 4, 5, 6, 112, 113, increases the fibril-forming tendency as seen in Table 7.
  • TABLE 7
    Tht fibrillation assay of lipidated pramlintide analogues
    Time taken to detect increase
    Peptide in fluorescence intensity (h)
    3 <5
    4 <5
    5 7
    6 15
    8 7
    9 7
    10 >144
    11 >144
    12 >144
    13 25
    15 >144
    17 >144
    18 >144
    19 >144
    20 >144
    21 >144
    24 >144
    25 >144
    28 45
    33 >144
    40 >144
    44 >144
    48 >144
    66 >144
    70 >144
    80 >144
    103 <5
    104 <5
    112 <5
    113 <5
    114 <5
    115 <5
    129 >144
    156 >144
  • Example 4 Pharmacokinetic Determination Via IV and SC Administration in Sprague Dawley Rats
  • The objective of the pharmacokinetic (PK) studies were to determine the plasma pharmacokinetic profile of lipidated pramlintide analogue peptides in fasted male SD rats after single intravenous (IV) and subcutaneous (SC) administration. PK studies were performed to determine the half-life (T1/2) of test peptides. T1/2 describes the time taken for the maximum plasma concentration (Cmax) of a test substance to halve its steady-state concentration when in circulation.
  • Male SD rats were purchased from Si Bei Fu Laboratory Animal Technology Co. Ltd (China). The animals were 6-8 weeks old with body weights of 200-300 g on the dosing date. The animals were housed in a 12-hour light/12-hour dark cycle environment and were fasted overnight before dosing. The body weight of the animals were recorded before dosing, 24 h and 48 h post dosing. Animals had free access to food and drinks, and the food consumption was quantified every day.
  • Test articles were administered at 20 nmol/kg. Blood samples were collected from each animal via Jugular vein. The sampling timepoints are as below.
  • Blood Samples per test article;
  • Group Route Animals Time points
    1 IV 1 2 3 0, 0.033, 0.1, 0.167, 0.25, 0.5,
    1, 2, 4, 6, 8, 12, 24, 48 h
    2 SC 4 5 6 0, 0.033, 0.1, 0.167, 0.25, 0.5,
    1, 2, 4 6, 8, 12, 24, 36, 48 h
  • The blood samples were transferred into eppendorf low binding tube containing K2EDTA. Above 0.150 mL blood were collected at each time point. Blood samples were centrifuged at 4,000 g for 5 minutes at 4° C. to obtain plasma. The plasma samples were stored frozen at −75±15° C. until analysis.
  • Concentrations of the test articles in the plasma samples were analyzed using a LC-MS/MS method. Data acquisition was performed by LabSolution version 5.89 software (Shimadzu, Kyoto, Japan). Data statistics were performed using Excel 97-2003 software. The pharmacokinetic parameters of test articles were calculated using a non-compartmental approach with Phoenix™ WinNonlin®6.1.
  • The following pharmacokinetic parameters were calculated, whenever possible from the plasma concentration versus time data:
      • IV administration: T1/2, C0, AUClast, AUCinf, MRTinf, Cl, Vss, Number of Points for Regression.
      • SC administration: Tmax, Cmax, AUClast, AUCinf, MRTinf, F, Number of Points for Regression.
  • TABLE 8
    Half-life of lipidated pramlintide analogues in rats
    Rat T1/2 (h)
    ID IV SC
    3 14
    4 12 12
    5 10.5 9.4
    6 9.5 17.6
    8 4.2 6.2
    9 5.9 7.8
    10 10.1 8.8
    12 9.2 9.7
    15 10.4 11.5
    18 12 13.6
    19 6.6 9.3
    20 10 10.8
    21 8 8.1
    24 9.4 10.5
    25 6.9 8
    35 5.4 8.1
    36 7.2 11.6
    38 6.6 9
    40 7.4 8.1
    41 8.3 10.1
    43 9.3 10
    44 8.9 11
    48 5.6 8.1
    64 6 5
    65 6 9.3
    66 9.4 12.2
    69 6.3 8.5
    70 10.3 12.5
    80 9.7 9.9
    112 11.8 19
    113 11.2 18.2
    114 13 21.6
    115 14.9 20.3
    117 9.8 12.9
    129 10.3 9
    140 10.6 12.9
    149 6.9 9.3
    155 7.7 8.9

    Pharmacokinetic studies show that the terminal half-life of amylin in rats is around 13 minutes, and the half-life for pramlintide in human is ˜20-45 minutes (Roth J D et. al. GLP-1R and amylin agonism in metabolic disease: complementary mechanisms and future opportunities. Br J Pharmacol. 2012; 166(1):121-136). The lipidated polypeptides show marked improvement in prolonging circulatory T½ compared to pramlintide.
  • Example 6 Rat Acute Food Intake Study
  • Male Sprague Dawley rats were obtained from Taconic Denmark, ApS at approximately 7 weeks of age. Rats were implanted with a microchip for identification, housed 4/cage with enrichment, free access to food and water, and allowed one week acclimatisation while non-invasive characterization was performed. Rats were on a 12:12 light:dark cycle that switches at 1 pm:1 am. Food intake was monitored via the HM2 system (Lafayette Instrument) that allows for monitoring in a home cage. As each rat enters an access tunnel to feed, an IR beam is broken, and the implanted microchip is read. Resulting changes to food weight is then assigned to the specific animal. Social order has shown no impact to overall feeding patterns and amounts.
  • Rats were sorted into groups based on Day −1 body weight and 24-hour accumulated food intake (n=7 per group). On Day 0 rats were weighed, then fasted for 6 hours. Thirty (30) minutes prior to the reintroduction of food, rats were dosed subcutaneously (5 mL/kg) with 20 nmol/kg of test compound or 60 nmol/kg peptide 1 (pramlintide) diluted in an appropriate vehicle, after which food was returned, and lights went out. Automated food intake was monitored for the following 3 days, and rats were weighed once per day.
  • Food intake per rat was batched into 1-hour intervals and integrated into Gubra's GubraView data management system. Discrete food intake data was exported into MS Excel from which cumulative food intake data was generated. Cumulative food intake data was then transposed into GraphPad Prism (v8.0.1) for analysis of dark period feeding
  • The lipidated polypeptides show marked suppression of food intake compared to pramlintide.
  • TABLE 9
    Effect of lipidated pramlintide
    analogues on food uptake in lean rats
    Cummulative food intake
    (% vehicle treated intake) )
    Peptide At 12 h At 24 h At 48 h
    1 71.6 85.6 98.7
    3 13.4 14.4 30.3
    8 28.1 42.7 68.8
    9 21.9 30.8 56.5
    10 18.8 24.6 46.6
    12 9.4 7.5 23.6
    15 45.2 49.1 51
    18 45.8 46.9 68.8
    20 27 26.2 49.7
    24 18.7 19.4 40.6
    35 49.8 44.8 72
    38 6.9 5.5 13.7
    40 60.7 51.8 72.4
    41 44.6 38.5 58.8
    43 60.8 55.6 46.6
    44 33.5 39.2 60.7
    48 27.5 32.6 57.1
    112 15.8 15.9 22.1
    113 69.5 65.8 77.9
    114 42.3 45.1 53.8
    115 42.4 41.3 52.2
    116 51.7 43.1 50.2
    129 29 32.4 51
    140 62.4 54.4 56.5
    149 31 31.8 17.5

Claims (53)

1. A polypeptide, or a pharmaceutically acceptable salt thereof, comprising the amino acid sequence:
Xaa (−4)-Xaa (−3)-Xaa (−2)-Xaa (−1)-Xaa 1-Cys 2-Asn 3-Xaa 4-Ala 5-Thr 6-Cys 7-Ala 8-Thr 9-Gln 10-Arg 11-Leu 12-Ala 13-Xaa 14-Xaa 15-Xaa 16-Xaa 17-His 18-Ser 19-Xaa 20-Xaa 21-Xaa 22-Xaa 23-Xaa 24-Xaa 25-Xaa 26-Xaa 27-Xaa 28-Xaa 29-Thr 30-Xaa 31 -Xaa 32-Xaa 33-Xaa 34-Xaa 35-Xaa 36-Xaa37-amide [SEQ ID NO:2], wherein:
Xaa (−4) is Lys(albumin binding moiety) or is absent;
Xaa (−3) is Gly or is absent;
Xaa (−2) is Gly or is absent;
Xaa (−1) is Gly, (albumin binding moiety), Lys(albumin binding moiety) or is absent;
Xaa 1 is Lys, Lys(albumin binding moiety), (albumin binding moiety) or is absent;
Xaa 4 is Thr, Ile or Ala;
Xaa 14 is Asn, His, Glu, 2,4-diaminobutanoic acid (Dab), or an alpha methyl amino acid;
Xaa 15 is Phe or Trp;
Xaa 16 is Leu or D-Leu (dL);
Xaa 17 is Val, Ser, Glu, Arg, (2S,4R)-4-hydroxypyrrolidine-2-carboxylic acid (Hyp), Dab or an alpha methyl amino acid (e.g. 2-amino-2-methylpropanoic acid [Aib]);
Xaa 20 is Ser, Ile, Pro or an alpha methyl amino acid (e.g. (S)-2-amino-3-hydroxy-2-methylpropanoic acid [αMeSer]);
Xaa 21 is Asn, Dab, His, Pro, Ser, Arg, Lys, Gly or Glu, Ala, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 22 is Asn, His, Hyp, Dab or an alpha methyl amino acid (e.g. Aib);
Xaa 23 is Phe, Hyp or an alpha methyl amino acid (e.g. (S)-2-amino-2-methyl-3-phenylpropanoic acid [αMePhe]);
Xaa 24 is Gly, Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 25 is Pro, Ala, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 26 is Ile, D-Ile (dI), Arg, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 27 is Leu, dL, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 28 is Pro, D-Pro (dP), Ser, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 29 is Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 31 is Asn, Glu, His, Arg, Pro, Dab or an alpha methyl amino acid (e.g. Aib);
Xaa 32 is Val, Hyp, Dab or an alpha methyl amino acid (e.g. Aib);
Xaa 33 is Gly, Pro, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 34 is Ser, Pro, His, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 35 is Asn, Pro, Arg, Glu, Dab, Hyp or an alpha methyl amino acid (e.g. Aib);
Xaa 36 is Thr, Hyp or an alpha methyl amino acid (e.g. Aib); and
Xaa 37 is Tyr, Pro, Hyp or an alpha methyl amino acid (e.g. Aib),
and wherein the polypeptide comprises at least one albumin binding moiety.
2. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein the polypeptide comprises Dab, Hyp or an alpha methyl amino acid at least one of positions 14, 17 or 20-37, optionally wherein the alpha methyl amino acid is Aib, αMePhe or αMeSer.
3. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 14 is Dab.
4. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 16 is dL.
5. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 17 is an alpha methyl amino acid, optionally Aib.
6. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 20 is an alpha methyl amino acid, optionally αMeS.
7. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 21 is an alpha methyl amino acid, optionally Aib.
8. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 21 is Dab.
9. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 22 is an alpha methyl amino acid, optionally Aib.
10. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 23 is an alpha methyl amino acid, optionally αMePhe.
11. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 24 is an alpha methyl amino acid, optionally Aib.
12. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 24 is Hyp.
13. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 25 is an alpha methyl amino acid, optionally Aib.
14. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 26 is an alpha methyl amino acid, optionally Aib.
15. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 26 is dl.
16. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 27 is an alpha methyl amino acid, optionally Aib.
17. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 27 is dL.
18. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 28 is an alpha methyl amino acid, optionally Aib.
19. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 28 is dP.
20. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 29 is an alpha methyl amino acid, optionally Aib.
21. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 31 is an alpha methyl amino acid, optionally Aib.
22. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 31 is Dab.
23. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 32 is an alpha methyl amino acid, optionally Aib.
24. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 33 is an alpha methyl amino acid, optionally Aib.
25. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 34 is an alpha methyl amino acid, optionally Aib.
26. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 35 is an alpha methyl amino acid, optionally Aib.
27. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 35 is Dab.
28. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 36 is an alpha methyl amino acid, optionally Aib.
29. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa 37 is an alpha methyl amino acid, optionally Aib.
30. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein the polypeptide comprises any one of the following combinations of amino acid modifications:
−1G, −2G, 17Aib
4A, 15W, 21P, 24P, 25A, 28S
4I, 20I, 21A, 35R
4I, 21Dab, 35R
14Dab
14Dab, 17Aib, 31E
14Dab, 23αMePhe, 21E
14Dab, 31E
14E, 17Aib
14E, 17Aib, 21H
14E, 17R
14E, 17R, 23αMePhe
14E, 21Aib
14H, 17Aib
14H, 17Aib, 21Aib, 31E
14H, 21Aib
14H, 21Aib, 35E
16dL, 21Aib
17Aib
17Aib, 37P
17Aib, 21Aib
17Aib, 21Aib, 37P
17Aib, 21G
17Aib, 21H
17Aib, 21K
17Aib, 21P
17Aib, 21P, 31E
17Aib, 21P, 35E
17Aib, 21R
17Aib, 21S
17Aib, 21Dab
17Aib, 21Dab, 31E
17Aib, 22H
17Aib, 22H, 35E
17Aib, 23αMePhe
17Aib, 26Aib
17Aib, 26R
17Aib, 27Aib
17Aib, 27dL
17Aib, 28Aib
17Aib, 29Aib
17Aib, 31Aib
17Aib, 31E
17Aib, 31H, 35E
17Aib, 31P
17Aib, 31R
17Aib, 32Aib
17Aib, 33Aib
17Aib, 34Aib
17Aib, 34H
17Aib, 34P
17Aib, 35Aib
17Aib, 35E
17Aib, 35R
17E, 21Aib
17R, 21Aib
17R, 21Aib, 31Aib
17R, 21Aib, 31E
17R, 21Aib, 31R
17R, 21Aib, 35Aib
17R, 23αMePhe
17R, 23αMePhe, 31E
17R, 26Aib
17S, 21Aib
17S, 21Aib, 31H
17S, 21Aib, 31P
17S, 21Aib, 31R
17S, 21Aib, 33P
17S, 21Aib, 35P
20αMeSer
20P, 21P, 24P, 25A, 28S
21Aib
21Aib, 24P, 25A, 28S
21Aib, 24P, 25A, 28S, 31Dab
21Aib, 24P, 25A, 28S, 35Dab
21Aib, 26dl
21Aib, 26Aib
21Aib, 27Aib
21Aib, 27dL
21Aib, 28Aib
21Aib, 28dP
21Aib, 31Aib
21Aib, 31E
21Aib, 31H
21Aib, 31R
21Aib, 33Aib
21Aib, 34Aib
21Aib, 35Aib
21Aib, 35E
21Aib, 35R
21Aib, 36Aib
21Aib, 37Aib
21Aib, 37P
21Dab, 24Hyp, 25A, 28S
21Dab, 24P, 25A, 28S
21Dab, 25Aib
21Dab, 31E
21P, 24P, 25A, 28S
22Aib
22H, 35E
23 αMePhe
23αMePhe, 31E
23αMePhe, 31R
23αMePhe, 35R
24Aib
26Aib
27Aib
27dL
28dP
35R
Δ1K, 4I, 21Dab, 35R
Δ1K, 14E, 17R, 23αMePhe.
31. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein:
Xaa (−1) is (albumin binding moiety), Lys(albumin binding moiety) or is absent;
Xaa 4 is Thr;
Xaa 14 is Asn, Glu or Dab;
Xaa 15 is Phe;
Xaa 16 is Leu;
Xaa 17 is Val, Arg or Aib;
Xaa 20 is Ser;
Xaa 21 is Asn—
Xaa 22 is Asn;
Xaa 23 is Phe or αMePhe;
Xaa 24 is Gly;
Xaa 25 is Pro;
Xaa 26 is Ile;
Xaa 27 is Leu;
Xaa 28 is Pro;
Xaa 29 is Pro;
Xaa 31 is Asn, Glu or Arg;
Xaa 32 is Val;
Xaa 33 is Gly;
Xaa 34 is Ser; and
Xaa 35 is Asn or Arg;
Xaa 36 is Thr; and
Xaa 37 is Tyr.
32-40. (canceled)
41. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein:
Xaa (−1) is Gly, Lys(albumin binding moiety) or (albumin binding moiety);
Xaa 4 is Thr;
Xaa 14 is Asn or His;
Xaa 15 is Phe;
Xaa 16 is Leu;
Xaa 17 is Val, Arg or Aib;
Xaa 20 is Ser;
Xaa 21 is Asn, Dab or Aib;
Xaa 22 is Asn;
Xaa 23 is Phe or αMePhe;
Xaa 24 is Gly;
Xaa 25 is Pro;
Xaa 26 is Ile or Aib;
Xaa 27 is Leu or Aib;
Xaa 28 is Pro or Aib;
Xaa 29 is Pro or Aib;
Xaa 31 is Asn, Glu or Aib;
Xaa 32 is Val or Aib;
Xaa 33 is Gly or Aib;
Xaa 34 is Ser or Aib;
Xaa 35 is Asn or Aib;
Xaa 36 is Thr or Aib; and
Xaa 37 is Tyr, Pro or Aib,
wherein the polypeptide comprises at least 2 Aib residues.
42-51. (canceled)
52. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein:
Xaa (−1) is Gly, Lys(albumin binding moiety), or (albumin binding moiety);
Xaa 14 is Asn, Glu or His;
Xaa 15 is Phe
Xaa 17 is Val, Ser, Glu, Arg or Aib;
Xaa 20 is Ser;
Xaa 21 is Asn or Aib;
Xaa 22 is Asn;
Xaa 23 is Phe;
Xaa 24 is Gly, Hyp or Pro;
Xaa 25 is Pro or Ala;
Xaa 26 is Ile, dl, or Aib;
Xaa 27 is Leu, dL, or Aib;
Xaa 28 is Pro, dP, Ser, or Aib;
Xaa 29 is Pro
Xaa 32 is Val;
Xaa 33 is Gly, Pro or Aib;
Xaa 34 is Ser or Aib;
Xaa 35 is Asn, Pro, Arg, Glu, Dab, or Aib;
Xaa 36 is Thr, or Aib; and
Xaa 37 is Tyr, Pro or Aib.
53-64. (canceled)
65. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein:
Xaa (−1) is (albumin binding moiety), Lys(albumin binding moiety), or is absent;
Xaa 4 is Thr;
Xaa 15 is Phe;
Xaa 16 is Leu;
Xaa 17 is Val or Aib;
Xaa 20 is Ser;
Xaa 21 is Asn, His, Pro, Ser, Arg, Dab, Lys or Gly;
Xaa 22 is Asn or His;
Xaa 23 is Phe;
Xaa 24 is Gly;
Xaa 25 is Pro;
Xaa 26 is Ile or Arg;
Xaa 27 is Leu or dL;
Xaa 28 is Pro;
Xaa 29 is Pro;
Xaa 31 is Asn, Glu, His, Arg or Pro;
Xaa 32 is Val;
Xaa 33 is Gly;
Xaa 34 is Ser, Pro or His;
Xaa 35 is Asn, Glu or Arg; Xaa 36 is Thr; and
Xaa 37 is Tyr.
66-69. (canceled)
70. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein Xaa (−4) is Lys(albumin binding moiety), Xaa (−1) is Lys(albumin binding moiety) or (albumin binding moiety), or Xaa 1 is Lys(albumin binding moiety).
71. (canceled)
72. (canceled)
73. The polypeptide or pharmaceutically acceptable salt of claim 1, wherein the albumin binding moiety is attached to an amino acid residue of the polypeptide.
74. (canceled)
75. (canceled)
76. A polypeptide selected from the group consisting of:
(SEQ ID NO: 3) C18diacid-γE- K[CNTATC]ATQRLANFLVHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 4) C18diacid-γE-γE-GGG- K[CNTATC]ATQRLANFLVHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 5) K(γE-γE-C18diacid) [CNTATC]ATQRLANFLVHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 6) K(O2Oc-O2Oc-γE-C18diacid) [CNTATC]ATQRLANFLVHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 7) K(O2Oc-O2Oc-γE-C18diacid) GGGK[CNTATC]ATQRLANFLVHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 8) K(γE-γE-C18diacid) [CNTATC]ATQRLANFLVHSS(Dab)NFPAILSPTNVGSNTY-amide (SEQ ID NO: 9) K(γE-γE-C18diacid) [CNTATC]ATQRLANFLVHSS(Aib)NFPAILSPTNVGSNTY-amide (SEQ ID NO: 10) C18diacid-γE-[CNTATC] ATQRLAEFLRHSSNN(αMePhe)GPILPPTNVGSNTY-amide (SEQ ID NO: 11) K(γE-γE-C18diacid)[CNTATC] ATQRLAEFLRHSSNN(αMePhe)GPILPPTNVGSNTY-amide (SEQ ID NO: 12) K(γE-C18diacid)K[CNTATC] ATQRLAEFLRHSSNN(αMePhe)GPILPPTNVGSNTY-amide (SEQ ID NO: 13) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSSNN(αMePhe)GPILPPTNVGSNTY-amide (SEQ ID NO: 14) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSSNN(αMePhe)GPILPPTNVGSNTY-amide (SEQ ID NO: 15) K(γE-C18diacid)K[CNTATC] ATQRLANFLRHSSNN(αMePhe)GPILPPTEVGSNTY-amide (SEQ ID NO: 16) K(γE-C18diacid)[CNTATC] ATQRLANFLRHSSNN(αMePhe)GPILPPTEVGSNTY-amide (SEQ ID NO: 17) K(O2Oc-O2Oc-γE-C18diacid)[CNTATC] ATQRLANFLRHSSNN(αMePhe)GPILPPTEVGSNTY-amide (SEQ ID NO: 18) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLRHSSNN(αMePhe)GPILPPTEVGSNTY-amide (SEQ ID NO: 19) K(γE-C18diacid)[CNTATC] ATQRLANFLRHSSNN(αMePhe)GPILPPTNVGSNTY-amide (SEQ ID NO: 20) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLRHSSNN(αMePhe)GPILPPTNVGSNTY-amide (SEQ ID NO: 21) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSSNN(αMePhe)GPILPPTNVGSRTY-amide (SEQ ID NO: 22) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHS(αMeSer)NNFGPILPPTNVGSNTY-amide (SEQ ID NO: 23) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSSNN(αMePhe)GPILPPTEVGSNTY-amide (SEQ ID NO: 24) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSSNN(αMePhe)GPILPPTRVGSNTY-amide (SEQ ID NO: 25) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSSNN(αMePhe)GPILPPTRVGSNTY-amide (SEQ ID NO: 26) K(γE-γE-C18diacid)[CNTATC] ATQRLANFL(Aib)HSSNN(αMePhe)GPILPPTNVGSNTY-amide (SEQ ID NO: 27) K(γE-γE-C18diacid)[CNIATC] ATQRLANFLVHSS(Dab)NFGPILPPTNVGSRTY-amide (SEQ ID NO: 28) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Dab)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 29) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Dab)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 30) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSS(Dab)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 31) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Dab)NFG(Aib)ILPPTNVGSNTY-amide (SEQ ID NO: 32) K(γE-C18diacid)K[CNTATC] ATQRLA(Dab)FLVHSSNN(αMePhe)GPILPPTEVGSNTY-amide (SEQ ID NO: 33) K(γE-C18diacid)K[CNTATC] ATQRLA(Dab)FLVHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 34) K(γE-C18diacid)K[CNTATC] ATQRLA(Dab)FLVHSSNNFGPILPPTEVGSNTY-amide (SEQ ID NO: 35) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 36) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 37) K(γE-C18diacid)K[CNTATC] ATQRLANFLSHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 38) K(γE-C18diacid)K[CNTATC] ATQRLAEFLVHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 39) K(γE-C18diacid)K[CNTATC] ATQRLANFLEHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 40) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTHVGSNTY-amide (SEQ ID NO: 41) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 42) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSETY-amide (SEQ ID NO: 43) K(γE-C18diacid)K[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 44) K(γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 45) K(O2Oc-O2Oc-γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 46) K(γE-C18diacid)K[CNTATC] ATQRLAHFLVHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 47) K(γE-C18diacid)K[CNTATC] ATQRLAHFLVHSS(Aib)NFGPILPPTNVGSETY-amide (SEQ ID NO: 48) K(γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 49) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 50) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 51) K(γE-C18diacid)K[CNTATC] ATQRLANFLSHSS(Aib)NFGPILPPTHVGSNTY-amide (SEQ ID NO: 52) K(γE-C18diacid)K[CNTATC] ATQRLANFLSHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 53) K(γE-C18diacid)K[CNTATC] ATQRLANFLSHSS(Aib)NFGPILPPTPVGSNTY-amide (SEQ ID NO: 54) K(γE-C18diacid)K[CNTATC] ATQRLANFLSHSS(Aib)NFGPILPPTNVPSNTY-amide (SEQ ID NO: 55) K(γE-C18diacid)K[CNTATC] ATQRLANFLSHSS(Aib)NFGPILPPTNVGSPTY-amide (SEQ ID NO: 56) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 57) K(γE-γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 58) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSNTP-amide (SEQ ID NO: 59) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPI(dL)PPTNVGSNTY-amide (SEQ ID NO: 60) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPIL(dP)PTNVGSNTY-amide (SEQ ID NO: 61) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGP(dI)LPPTNVGSNTY-amide (SEQ ID NO: 62) K(γE-C18diacid)[CNTATC] ATQRLANF(dL)VHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 63) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 64) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSRTY-amide (SEQ ID NO: 65) K(γE-C18diacid)K[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 66) K(γE-γE-C18diacid)K[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 67) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 68) (C18diacid-γE-[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 69) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 70) K(γE-γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 71) C18diacid-γE-K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 72) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSRTY-amide (SEQ ID NO: 73) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSRTY-amide (SEQ ID NO: 74) K(γE-γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSRTY-amide (SEQ ID NO: 75) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 76) K(γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 77) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTRVGSNTY-amide (SEQ ID NO: 78) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGP(Aib)LPPTNVGSNTY-amide (SEQ ID NO: 79) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPI(Aib)PPTNVGSNTY-amide (SEQ ID NO: 80) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPT(Aib)VGSNTY-amide (SEQ ID NO: 81) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNV(Aib)SNTY-amide (SEQ ID NO: 82) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGS(Aib)TY-amide (SEQ ID NO: 83) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSN(Aib)Y-amide (SEQ ID NO: 84) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVG(Aib)NTY-amide (SEQ ID NO: 85) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGSNT(Aib)-amide (SEQ ID NO: 86) K(γE-C18diacid)K[CNTATC] ATQRLAHFL(Aib)HSS(Aib)NFGPILPPTEVGSNTY-amide (SEQ ID NO: 87) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSS(Aib)NFGPILPPTNVGSNTP-amide (SEQ ID NO: 88) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPIL(Aib)PTNVGSNTY-amide (SEQ ID NO: 89) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPT(Aib)VGSNTY-amide (SEQ ID NO: 90) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPT(Aib)VGSNTY-amide (SEQ ID NO: 91) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPTNVGS(Aib)TY-amide (SEQ ID NO: 92) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLRHSS(Aib)NFGPILPPTNVGS(Aib)TY-amide (SEQ ID NO: 93) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSS(Dab)NFG(Aib)ILPPTNVGSNTY-amide (SEQ ID NO: 94) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGP(Aib)LPPTNVGSNTY-amide (SEQ ID NO: 95) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPI(Aib)PPTNVGSNTY-amide (SEQ ID NO: 96) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPIL(Aib)PTNVGSNTY-amide (SEQ ID NO: 97) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILP(Aib)TNVGSNTY-amide (SEQ ID NO: 98) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPT(Aib)VGSNTY-amide (SEQ ID NO: 99) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTN(Aib)GSNTY-amide (SEQ ID NO: 100) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNV(Aib)SNTY-amide (SEQ ID NO: 101) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVG(Aib)NTY-amide (SEQ ID NO: 102) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGS(Aib)TY-amide (SEQ ID NO: 103) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSSNNFGPI(dL)PPTNVGSNTY-amide (SEQ ID NO: 104) K(γE-C18diacid)[CNTATC] ATQRLANFLVHSSNNFGPIL(dP)PTNVGSNTY-amide (SEQ ID NO: 105) K(γE-C18diacid)K[CNTATC] ATQRLANFLSHSS(Dab)NFG(Aib)ILPPTNVGSNTY-amide (SEQ ID NO: 106) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSSNNFGP(Aib)LPPTNVGSNTY-amide (SEQ ID NO: 107) K(γE-C18diacid)K[CNTATC] ATQRLANFLRHSSNNFGP(Aib)LPPTNVGSNTY-amide (SEQ ID NO: 108) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSSNNFGPI(Aib)PPTNVGSNTY-amide (SEQ ID NO: 109) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSSN(Aib)FGPILPPTNVGSNTY-amide (SEQ ID NO: 110) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSSNNF(Aib)PILPPTNVGSNTY-amide (SEQ ID NO: 111) K(γE-C18diacid)K[CNTATC] ATQRLANFLVHSSNHFGPILPPTNVGSETY-amide (SEQ ID NO: 112) C20diacid-γE-K[CNTATC] ATQRLANFLVHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 113) C2Odiacid-γE-O2Oc-O2Oc-K[CNTATC] ATQRLANFLVHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 114) K(γE-γE-C20diacid)[CNTATC] ATQRLANFLVHSSNNFGPILPPTNVGSRTY-amide (SEQ ID NO: 115) K(γE-γE-C20diacid)[CNTATC] ATQRLANFLVHSSPNFPAILSPTNVGSNTY-amide (SEQ ID NO: 116) K(γE-γE-C20diacid)[CNTATC] ATQRLAEFLRHSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 117) K(γE-γE-C20diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFPAILSPTNVGSNTY-amide (SEQ ID NO: 118) K(γE-γE-C20diacid)[CNIATC] ATQRLANFLVH5IANFGPILPPTNVGSRTY-amide (SEQ ID NO: 119) K(γE-γE-C20diacid)[CNTATC] ATQRLANFLVHSPPNFPAILSPTNVGSNTY-amide (SEQ ID NO: 120) K(γE-γE-C20diacid)[CNAATC] ATQRLANWLVHSSPNFPAILSPTNVGSNTY-amide (SEQ ID NO: 8) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Dab)NFPAILSPTNVGSNTY-amide (SEQ ID NO: 121) K(γE-γE-C20diacid)[CNTATC] ATQRLANFLVHSS(Aib)NF(Hyp)AILSPTNVGSNTY-amide (SEQ ID NO: 122) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFPAILSPT(Dab)VGSNTY-amide (SEQ ID NO: 123) K(γE-γE-C20diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFPAILSPT(Dab)VGSNTY-amide (SEQ ID NO: 124) K(γE-γE-C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFPAILSPTNVGS(Dab)TY-amide (SEQ ID NO: 125) K(γE-γE-C20diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFPAILSPTNVGS(Dab)TY-amide (SEQ ID NO: 126) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSS(Dab)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 127) K(γE-γE-C18diacid)[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 128) K(γE-C18diacid)K[CNTATC] ATQRLAEFL(Aib)HSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 129) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 130) K(γE-C18diacid)K[CNTATC] ATQRLAEFL(Aib)HSSHNFGPILPPTNVGSNTY-amide (SEQ ID NO: 131) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSHNFGPILPPTNVGSNTY-amide (SEQ ID NO: 132) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSPNFGPILPPTNVGSNTY-amide (SEQ ID NO: 133) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSSNFGPILPPTNVGSNTY-amide (SEQ ID NO: 134) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTPVGSNTY-amide (SEQ ID NO: 135) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNHFGPILPPTNVGSNTY-amide (SEQ ID NO: 136) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGSNTP-amide (SEQ ID NO: 137) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSRNFGPILPPTNVGSNTY-amide (SEQ ID NO: 138) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSPNFGPILPPTEVGSNTY-amide (SEQ ID NO: 139) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSPNFGPILPPTNVGSETY-amide (SEQ ID NO: 140) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTEVGSNTY-amide (SEQ ID NO: 141) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGSRTY-amide (SEQ ID NO: 142) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGSETY-amide (SEQ ID NO: 143) K(γE-C18diacid)GGK[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 144) K(γE-C18diacid)[CNTATC] ATQRLAHFL(Aib)HSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 145) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGHNTY-amide (SEQ ID NO: 146) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTHVGSETY-amide (SEQ ID NO: 147) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNHFGPILPPTNVGSETY-amide (SEQ ID NO: 148) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGPNTY-amide (SEQ ID NO: 149) K(γE-C18diacid)[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTNVGSNTY-amide (SEQ ID NO: 150) K(γE-C18diacid)[CNTATC] ATQRLANFL(Aib)HSS(Dab)NFGPILPPTNVGSNTY-amide (SEQ ID NO: 151) K(γE-C18diacid)[CNTATC] ATQRLANFL(Aib)HSSNNFGPI(dL)PPTNVGSNTY-amide (SEQ ID NO: 152) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSNNFGPRLPPTNVGSNTY-amide (SEQ ID NO: 153) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSKNFGPILPPTNVGSNTY-amide (SEQ ID NO: 154) K(γE-C18diacid)K[CNTATC] ATQRLANFL(Aib)HSSGNFGPILPPTNVGSNTY-amide (SEQ ID NO: 155) K(γE-C18diacid)[CNTATC] ATQRLANFL(Aib)HSSNNFGPILPPTRVGSNTY-amide (SEQ ID NO: 156) K(γE-C18diacid)K[CNTATC] ATQRLA(Dab)FL(Aib)HSSNNFGPILPPTEVGSNTY-amide (SEQ ID NO: 157) (C18diacid)K[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPT(Aib)VGSNTY-amide (SEQ ID NO: 158) K(C18diacid)[CNTATC] ATQRLANFLVHSS(Aib)NFGPILPPT(Aib)VGSNTY-amide
77. A pharmaceutical composition comprising the polypeptide or pharmaceutically acceptable salt of claim 1 and a pharmaceutically acceptable excipient.
78. A method for treating and/or preventing a disease or disorder in a subject comprising administering the polypeptide or pharmaceutically acceptable salt of claim 1 or the pharmaceutical composition of claim 77.
79. The method of claim 78, wherein the disease or disorder is obesity, metabolic disease, an obesity-related condition, eating disorder, Alzheimer's disease, hepatic steatosis (“fatty liver”), kidney failure, arteriosclerosis (e.g. atherosclerosis), cardiovascular disease, macrovascular disease, microvascular disease, diabetic heart (including diabetic cardiomyopathy and heart failure as a diabetic complication), coronary heart disease, peripheral artery disease or stroke, cancer, dumping syndrome, hypertension e.g. pulmonary hypertension, or dyslipidemia e.g. atherogenic dyslipidemia, cholescystitis, or short bowel syndrome.
80-83. (canceled)
84. A method for the production of the polypeptide of claim 1.
85. (canceled)
86. An article of manufacture comprising the polypeptide or pharmaceutically acceptable salt of claim 1 or the pharmaceutical composition of claim 77.
87. A kit comprising the polypeptide or pharmaceutically acceptable salt of claim 1 or the pharmaceutical composition of claim 77, optionally further comprising instructions for use.
US17/551,701 2020-12-16 2021-12-15 Polypeptides and uses thereof Pending US20220204580A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/551,701 US20220204580A1 (en) 2020-12-16 2021-12-15 Polypeptides and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063125996P 2020-12-16 2020-12-16
US17/551,701 US20220204580A1 (en) 2020-12-16 2021-12-15 Polypeptides and uses thereof

Publications (1)

Publication Number Publication Date
US20220204580A1 true US20220204580A1 (en) 2022-06-30

Family

ID=79287870

Family Applications (2)

Application Number Title Priority Date Filing Date
US18/257,756 Pending US20240067693A1 (en) 2020-12-16 2021-12-15 Polypeptides and uses thereof
US17/551,701 Pending US20220204580A1 (en) 2020-12-16 2021-12-15 Polypeptides and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US18/257,756 Pending US20240067693A1 (en) 2020-12-16 2021-12-15 Polypeptides and uses thereof

Country Status (16)

Country Link
US (2) US20240067693A1 (en)
EP (1) EP4263586A1 (en)
JP (1) JP2023554046A (en)
KR (1) KR20230120134A (en)
CN (1) CN116685599A (en)
AR (1) AR124391A1 (en)
AU (1) AU2021399076A1 (en)
CA (1) CA3204296A1 (en)
CL (1) CL2023001719A1 (en)
CO (1) CO2023009066A2 (en)
CR (1) CR20230311A (en)
EC (1) ECSP23052600A (en)
IL (1) IL303578A (en)
MX (1) MX2023007288A (en)
TW (1) TW202241933A (en)
WO (1) WO2022129254A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11744873B2 (en) 2021-01-20 2023-09-05 Viking Therapeutics, Inc. Compositions and methods for the treatment of metabolic and liver disorders

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101400698A (en) * 2006-03-15 2009-04-01 诺沃-诺迪斯克有限公司 Amylin derivatives
EP2036923A1 (en) * 2007-09-11 2009-03-18 Novo Nordisk A/S Improved derivates of amylin
WO2010046357A1 (en) * 2008-10-21 2010-04-29 Novo Nordisk A/S Amylin derivatives
WO2013059336A1 (en) * 2011-10-18 2013-04-25 Amylin Pharmaceuticals, Llc Amylin-calcitonin chimeric peptides conjugated to duration enchancing moieties
ES2739289T3 (en) * 2014-11-27 2020-01-30 Novo Nordisk As GLP-1 derivatives and their uses
WO2020225781A1 (en) * 2019-05-08 2020-11-12 Auckland Uniservices Limited Peptide conjugate amylin agonists and uses thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11744873B2 (en) 2021-01-20 2023-09-05 Viking Therapeutics, Inc. Compositions and methods for the treatment of metabolic and liver disorders

Also Published As

Publication number Publication date
KR20230120134A (en) 2023-08-16
TW202241933A (en) 2022-11-01
AR124391A1 (en) 2023-03-22
MX2023007288A (en) 2023-07-03
ECSP23052600A (en) 2023-08-31
US20240067693A1 (en) 2024-02-29
CL2023001719A1 (en) 2024-01-26
CR20230311A (en) 2023-09-01
AU2021399076A1 (en) 2023-07-27
JP2023554046A (en) 2023-12-26
IL303578A (en) 2023-08-01
WO2022129254A1 (en) 2022-06-23
EP4263586A1 (en) 2023-10-25
CO2023009066A2 (en) 2023-07-10
CA3204296A1 (en) 2022-06-23
CN116685599A (en) 2023-09-01

Similar Documents

Publication Publication Date Title
US11542313B2 (en) Incretin analogs and uses thereof
KR102440323B1 (en) Incretin analogues and uses thereof
EP2970511B1 (en) Insulin-incretin conjugates
US8343914B2 (en) Fibrillation resistant proteins
EP3206710B1 (en) Incretin-insulin conjugates
AU2003208945B2 (en) Method for administering GLP-1 molecules
US8993516B2 (en) Meal-time insulin analogues of enhanced stability
US20160058881A1 (en) Prodrugs with prolonged action
KR20120123443A (en) Glucagon antagonist - gip agonist conjugates and composition for the treatment of metabolic disorders and obesity
KR20110110174A (en) Amide based glucagon superfamily peptide prodrugs
US20160303245A1 (en) Fatty acid derivatives of dimeric inhibitors of psd-95
AU2020408139B2 (en) Incretin analogs and uses thereof
US20110152183A1 (en) Derivatised hybrid peptides of amylin and salmon calcitonin
US20220204580A1 (en) Polypeptides and uses thereof
US20240024425A1 (en) Pharmaceutical composition of glp-1/glp-2 dual agonists

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: MEDIMMUNE LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BEDNAREK, MARIA ALEKSANDRA;GENAPATHY, SIVANESWARY;SIGNING DATES FROM 20221017 TO 20221018;REEL/FRAME:061715/0642

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION RETURNED BACK TO PREEXAM

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION